1
|
Seixas MLGA, Mitre LP, Shams S, Lanzuolo GB, Bartolomeo CS, Silva EA, Prado CM, Ureshino R, Stilhano RS. Unraveling Muscle Impairment Associated With COVID-19 and the Role of 3D Culture in Its Investigation. Front Nutr 2022; 9:825629. [PMID: 35223956 PMCID: PMC8867096 DOI: 10.3389/fnut.2022.825629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been considered a public health emergency, extensively investigated by researchers. Accordingly, the respiratory tract has been the main research focus, with some other studies outlining the effects on the neurological, cardiovascular, and renal systems. However, concerning SARS-CoV-2 outcomes on skeletal muscle, scientific evidence is still not sufficiently strong to trace, treat and prevent possible muscle impairment due to the COVID-19. Simultaneously, there has been a considerable amount of studies reporting skeletal muscle damage in the context of COVID-19. Among the detrimental musculoskeletal conditions associated with the viral infection, the most commonly described are sarcopenia, cachexia, myalgia, myositis, rhabdomyolysis, atrophy, peripheral neuropathy, and Guillain-Barré Syndrome. Of note, the risk of developing sarcopenia during or after COVID-19 is relatively high, which poses special importance to the condition amid the SARS-CoV-2 infection. The yet uncovered mechanisms by which musculoskeletal injury takes place in COVID-19 and the lack of published methods tailored to study the correlation between COVID-19 and skeletal muscle hinder the ability of healthcare professionals to provide SARS-CoV-2 infected patients with an adequate treatment plan. The present review aims to minimize this burden by both thoroughly exploring the interaction between COVID-19 and the musculoskeletal system and examining the cutting-edge 3D cell culture techniques capable of revolutionizing the study of muscle dynamics.
Collapse
Affiliation(s)
- Maria Luiza G. A. Seixas
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Lucas Pari Mitre
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Shahin Shams
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Gabriel Barbugian Lanzuolo
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Cynthia Silva Bartolomeo
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
- Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
| | - Eduardo A. Silva
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Carla Maximo Prado
- Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo Ureshino
- Department of Biological Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Roberta Sessa Stilhano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
- *Correspondence: Roberta Sessa Stilhano
| |
Collapse
|
2
|
Deviatkin AA, Vakulenko YA, Akhmadishina LV, Tarasov VV, Beloukhova MI, Zamyatnin Jr. AA, Lukashev AN. Emerging Concepts and Challenges in Rheumatoid Arthritis Gene Therapy. Biomedicines 2020; 8:biomedicines8010009. [PMID: 31936504 PMCID: PMC7168286 DOI: 10.3390/biomedicines8010009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory joint disease affecting about 1% of the population worldwide. Current treatment approaches do not ensure a cure for every patient. Moreover, classical regimens are based on nontargeted systemic immune suppression and have significant side effects. Biological treatment has advanced considerably but efficacy and specificity issues remain. Gene therapy is one of the potential future directions for RA therapy, which is rapidly developing. Several gene therapy trials done so far have been of moderate success, but experimental and genetics studies have yielded novel targets. As a result, the arsenal of gene therapy tools keeps growing. Currently, both viral and nonviral delivery systems are used for RA therapy. Herein, we review recent approaches for RA gene therapy.
Collapse
Affiliation(s)
- Andrei A. Deviatkin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Correspondence:
| | - Yulia A. Vakulenko
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ludmila V. Akhmadishina
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| | - Vadim V. Tarasov
- Department of Pharmacology and Pharmacy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Marina I. Beloukhova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
| | - Andrey A. Zamyatnin Jr.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alexander N. Lukashev
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| |
Collapse
|
3
|
Bujold M, Caron N, Camiran G, Mukherjee S, Allen PD, Tremblay JP, Wang Y. Autotransplantation in mdx Mice of mdx Myoblasts Genetically Corrected by an HSV-1 Amplicon Vector. Cell Transplant 2017. [DOI: 10.3727/000000002783985297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder, characterized by a lack of dystrophin. To eliminate the need for immunosuppressive drugs, transplantation of genetically modified autologous myoblasts has been proposed as a possible therapy for this myopathy. An HSV-1 amplicon vector (HSVDGN), containing a 17.3-kb full-length MCK-driven mouse dystrophin cDNA, an eGFP gene, and a neomycin resistance gene driven by CMV or SV40 promoters, respectively, was constructed and used to transduce mdx primary myoblasts. The presence of the eGFP and neomycin resistance genes facilitated the evaluation of the initial transduction efficiency and the permanent transduction frequency. At low multiplicities of infection (MOI 1–5), the majority of myoblasts (60–90%) expressed GFP. The GFP-positive mdx myoblasts were sorted by FACS and selected with neomycin (300 μg/ml) for 2 weeks. Up to 2% of initially infected mdx myoblasts stably expressed the three transgenes without further selection at that time. These altered cells were grafted into the tibialis anterior muscles of 18 mdx mice. Some of the mice were immunosuppressed with FK506 due to the anticipation that eGFP and the product of neomycin resistance gene might be immunogenic. One month after transplantation, numerous muscle fibers expressing mouse dystrophin were detected by immunohistochemistry, in both immunosuppressed (10–50%) and nonimmunosuppressed (5–25%) mdx mice. Our results demonstrated the capability of permanently expressing a full-length dystrophin in dystrophic myoblasts with HSV-1 amplicon vector and raised the possibility of an eventual treatment of DMD based on the transplantation of genetically modified autologous myoblasts.
Collapse
Affiliation(s)
- Mathieu Bujold
- Laboratoire de Génétique Humaine, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL), Ste-Foy (Qc), Canada, G1V 4G2
| | - Nicolas Caron
- Laboratoire de Génétique Humaine, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL), Ste-Foy (Qc), Canada, G1V 4G2
| | - Goeffrey Camiran
- Laboratoire de Génétique Humaine, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL), Ste-Foy (Qc), Canada, G1V 4G2
| | | | - Paul. D. Allen
- Department of Anesthesia, Brigham & Women's Hospital, Boston, MA
| | - Jacques P. Tremblay
- Laboratoire de Génétique Humaine, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL), Ste-Foy (Qc), Canada, G1V 4G2
| | - Yaming Wang
- Department of Anesthesia, Brigham & Women's Hospital, Boston, MA
| |
Collapse
|
4
|
Viral Vector-Mediated Antisense Therapy for Genetic Diseases. Genes (Basel) 2017; 8:genes8020051. [PMID: 28134780 PMCID: PMC5333040 DOI: 10.3390/genes8020051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 01/16/2023] Open
Abstract
RNA plays complex roles in normal health and disease and is becoming an important target for therapeutic intervention; accordingly, therapeutic strategies that modulate RNA function have gained great interest over the past decade. Antisense oligonucleotides (AOs) are perhaps the most promising strategy to modulate RNA expression through a variety of post binding events such as gene silencing through degradative or non-degradative mechanisms, or splicing modulation which has recently demonstrated promising results. However, AO technology still faces issues like poor cellular-uptake, low efficacy in target tissues and relatively rapid clearance from the circulation which means repeated injections are essential to complete therapeutic efficacy. To overcome these limitations, viral vectors encoding small nuclear RNAs have been engineered to shuttle antisense sequences into cells, allowing appropriate subcellular localization with pre-mRNAs and permanent correction. In this review, we outline the different strategies for antisense therapy mediated by viral vectors and provide examples of each approach. We also address the advantages and limitations of viral vector use, with an emphasis on their clinical application.
Collapse
|
5
|
Lukashev AN, Zamyatnin AA. Viral Vectors for Gene Therapy: Current State and Clinical Perspectives. BIOCHEMISTRY (MOSCOW) 2017; 81:700-8. [PMID: 27449616 DOI: 10.1134/s0006297916070063] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Gene therapy is the straightforward approach for the application of recent advances in molecular biology into clinical practice. One of the major obstacles in the development of gene therapy is the delivery of the effector to and into the target cell. Unfortunately, most methods commonly used in laboratory practice are poorly suited for clinical use. Viral vectors are one of the most promising methods for gene therapy delivery. Millions of years of evolution of viruses have resulted in the development of various molecular mechanisms for entry into cells, long-term survival within cells, and activation, inhibition, or modification of the host defense mechanisms at all levels. The relatively simple organization of viruses, small genome size, and evolutionary plasticity allow modifying them to create effective instruments for gene therapy approaches. This review summarizes the latest trends in the development of gene therapy, in particular, various aspects and prospects of the development of clinical products based on viral delivery systems.
Collapse
Affiliation(s)
- A N Lukashev
- Chumakov Institute of Poliomyelitis and Viral Encephalitides, Moscow, 142782, Russia.
| | | |
Collapse
|
6
|
Co-activator binding protein PIMT mediates TNF-α induced insulin resistance in skeletal muscle via the transcriptional down-regulation of MEF2A and GLUT4. Sci Rep 2015; 5:15197. [PMID: 26468734 PMCID: PMC4606566 DOI: 10.1038/srep15197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/21/2015] [Indexed: 12/18/2022] Open
Abstract
The mechanisms underlying inflammation induced insulin resistance are poorly understood. Here, we report that the expression of PIMT, a transcriptional co-activator binding protein, was up-regulated in the soleus muscle of high sucrose diet (HSD) induced insulin resistant rats and TNF-α exposed cultured myoblasts. Moreover, TNF-α induced phosphorylation of PIMT at the ERK1/2 target site Ser298. Wild type (WT) PIMT or phospho-mimic Ser298Asp mutant but not phospho-deficient Ser298Ala PIMT mutant abrogated insulin stimulated glucose uptake by L6 myotubes and neonatal rat skeletal myoblasts. Whereas, PIMT knock down relieved TNF-α inhibited insulin signaling. Mechanistic analysis revealed that PIMT differentially regulated the expression of GLUT4, MEF2A, PGC-1α and HDAC5 in cultured cells and skeletal muscle of Wistar rats. Further characterization showed that PIMT was recruited to GLUT4, MEF2A and HDAC5 promoters and overexpression of PIMT abolished the activity of WT but not MEF2A binding defective mutant GLUT4 promoter. Collectively, we conclude that PIMT mediates TNF-α induced insulin resistance at the skeletal muscle via the transcriptional modulation of GLUT4, MEF2A, PGC-1α and HDAC5 genes.
Collapse
|
7
|
McFall ER, Murray LM, Lunde JA, Jasmin BJ, Kothary R, Parks RJ. A reduction in the human adenovirus virion size through use of a shortened fibre protein does not enhance muscle transduction following systemic or localised delivery in mice. Virology 2014; 468-470:444-453. [PMID: 25243333 DOI: 10.1016/j.virol.2014.08.026] [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: 06/17/2014] [Revised: 07/28/2014] [Accepted: 08/22/2014] [Indexed: 02/06/2023]
Abstract
We have investigated whether reducing the overall size of adenovirus (Ad), through use of a vector containing a shortened fibre, leads to enhanced distribution and dissemination of the vector. Intravenous or intraperitoneal injection of Ad5SlacZ (12 nm fibre versus the normal Ad5 37 nm fibre) or Ad5SpKlacZ (shortened fibre with polylysine motif in the H-I loop of fibre knob domain) led to similar levels of lacZ expression compared to Ad5LlacZ (native Ad5 fibre) in the liver of treated animals, but did not enhance extravasation into the tibialis anterior muscle. Direct injection of the short-fibre vectors into the tibialis anterior muscle did not result in enhanced spread of the vector through muscle tissue, and led to only sporadic transgene expression in the spinal cord, suggesting that modifying the fibre length or redirecting viral infection to a more common cell surface receptor does not enhance motor neuron uptake or retrograde transport.
Collapse
Affiliation(s)
- Emily R McFall
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Lyndsay M Murray
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6
| | - John A Lunde
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Ontario, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Robin J Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Ontario, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
8
|
Guitart M, Osorio-Conles Ó, Pentinat T, Cebrià J, García-Villoria J, Sala D, Sebastián D, Zorzano A, Ribes A, Jiménez-Chillarón JC, García-Martínez C, Gómez-Foix AM. Fatty acid transport protein 1 (FATP1) localizes in mitochondria in mouse skeletal muscle and regulates lipid and ketone body disposal. PLoS One 2014; 9:e98109. [PMID: 24858472 PMCID: PMC4032244 DOI: 10.1371/journal.pone.0098109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 04/29/2014] [Indexed: 12/20/2022] Open
Abstract
FATP1 mediates skeletal muscle cell fatty acid import, yet its intracellular localization and metabolic control role are not completely defined. Here, we examine FATP1 localization and metabolic effects of its overexpression in mouse skeletal muscle. The FATP1 protein was detected in mitochondrial and plasma membrane fractions, obtained by differential centrifugation, of mouse gastrocnemius muscle. FATP1 was most abundant in purified mitochondria, and in the outer membrane and soluble intermembrane, but not in the inner membrane plus matrix, enriched subfractions of purified mitochondria. Immunogold electron microscopy localized FATP1-GFP in mitochondria of transfected C2C12 myotubes. FATP1 was overexpressed in gastrocnemius mouse muscle, by adenovirus-mediated delivery of the gene into hindlimb muscles of newborn mice, fed after weaning a chow or high-fat diet. Compared to GFP delivery, FATP1 did not alter body weight, serum fed glucose, insulin and triglyceride levels, and whole-body glucose tolerance, in either diet. However, fatty acid levels were lower and β-hydroxybutyrate levels were higher in FATP1- than GFP-mice, irrespective of diet. Moreover, intramuscular triglyceride content was lower in FATP1- versus GFP-mice regardless of diet, and β-hydroxybutyrate content was unchanged in high-fat-fed mice. Electroporation-mediated FATP1 overexpression enhanced palmitate oxidation to CO2, but not to acid-soluble intermediate metabolites, while CO2 production from β-hydroxybutyrate was inhibited and that from glucose unchanged, in isolated mouse gastrocnemius strips. In summary, FATP1 was localized in mitochondria, in the outer membrane and intermembrane parts, of mouse skeletal muscle, what may be crucial for its metabolic effects. Overexpressed FATP1 enhanced disposal of both systemic fatty acids and intramuscular triglycerides. Consistently, it did not contribute to the high-fat diet-induced metabolic dysregulation. However, FATP1 lead to hyperketonemia, likely secondary to the sparing of ketone body oxidation by the enhanced oxidation of fatty acids.
Collapse
Affiliation(s)
- Maria Guitart
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- * E-mail:
| | - Óscar Osorio-Conles
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Thais Pentinat
- Hospital Sant Joan de Déu, Endocrinology, Esplugues, Barcelona, Spain
| | - Judith Cebrià
- Hospital Sant Joan de Déu, Endocrinology, Esplugues, Barcelona, Spain
| | - Judit García-Villoria
- Sección de Errores Congénitos del Metabolismo (IBC), Servicio de Bioquímica y Genética Molecular, Hospital Clínico, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain
| | - David Sala
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - David Sebastián
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Antonio Zorzano
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Antonia Ribes
- Sección de Errores Congénitos del Metabolismo (IBC), Servicio de Bioquímica y Genética Molecular, Hospital Clínico, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain
| | | | - Celia García-Martínez
- Departament de Patologia i Terapèutica Experimental, UB, Hospitalet de Llobregat, Barcelona, Spain
| | - Anna M. Gómez-Foix
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona (UB), Institut de Biomedicina de la UB, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| |
Collapse
|
9
|
Rivera-Meza M, Quintanilla ME, Tampier L. Reduction of ethanol consumption in alcohol-preferring rats by dual expression gene transfer. Alcohol Alcohol 2012; 47:102-8. [PMID: 22214999 DOI: 10.1093/alcalc/agr161] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS To mimic, in an animal model of alcoholism, the protective phenotype against alcohol consumption observed in humans carrying a fast alcohol dehydrogenase (ADH1B*2) and an inactive aldehyde dehydrogenase (ALDH2*2). METHODS We developed a multiple expression cassette adenoviral vector (AdV-ADH/asALDH2) encoding both a fast rat ADH and an antisense RNA against rat ALDH2. A control adenoviral vector (AdV-C) containing intronic non-coding DNA was also developed. These adenoviral vectors were administered intravenously to rats bred as high alcohol-drinkers (University of Chile bibulous) that were previously rendered alcohol dependent by a 75-day period of voluntary 10% ethanol intake. RESULTS Animals administered AdV-ADH/asALDH2 showed a 176% increase in liver ADH activity, whereas liver ALDH2 activity was reduced by 24%, and upon the administration of a dose of ethanol (1 g/kg, i.p.), these showed arterial acetaldehyde levels that were 400% higher than those of animals administered AdV-C. Rats that received the AdV-ADH/asALDH2 vector reduced by 60% their voluntary ethanol intake versus controls. CONCLUSION This study provides evidence that the simultaneous increase of liver ADH and a reduction of ALDH activity by gene transfer could constitute a potential therapeutic strategy for the treatment of alcoholism.
Collapse
Affiliation(s)
- Mario Rivera-Meza
- Faculty of Medicine, Institute of Biomedical Sciences, Program of Molecular and Clinical Pharmacology, Laboratory of Pharmacogenetics of Alcoholism, University of Chile, Av. Independencia 1027, Independencia, Santiago, Chile.
| | | | | |
Collapse
|
10
|
Parrish E, Peltékian E, Dickson G, Epstein AL, Garcia L. Cell engineering for muscle gene therapy: Extemporaneous production of retroviral vector packaging macrophages using defective herpes simplex virus type 1 vectors harbouring gag, pol, env genes. Cytotechnology 2011; 30:173-80. [PMID: 19003367 DOI: 10.1023/a:1008022713466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Gene therapy as a treatment for neuromuscular diseases is an ever-developing concept based on the use of DNA as the therapeutic agent. In the search for appropriate strategies a bottleneck exists, however, concerning the targeting of vectors carrying the therapeutic gene, to all pathologic sites. These diseases are often characterised by multiple widespread lesions spread over a large area, rendering administration by local injection into tissues, clinically irrelevant. With this in mind, we have proposed that circulating cells (monocytes/macrophages), which home naturally to inflammatory lesions, characteristic of degenerating muscle, could be used as shuttles able to track down every damaged site, and deliver there a corrective gene. Our aim is to mobilise a corrective gene from these infiltrating monocyte-macrophages, into muscle cells, a process of in situ cell to cell gene transfer which could be accomplished using a retroviral vector, since the regeneration process involves the proliferation of muscle precursors before they fuse to form replacement fibres. For this, monocyte-macrophages must be engineered into 'packaging cells' containing both the replication deficient retrovirus carrying the gene of interest and an helper genome (gag-pol-env) needed for its assembly and secretion. Here, we have transduced a monocyte cell line using a defective murine Moloney leukemia retrovirus carrying the LacZ reporter gene. This provided us with a platform to investigate the possibility of gag-pol-env vector driven packaging of the defective retrovirus by macrophages. We show that an herpes simplex virus type I amplicon harbouring the Moloney gag, pol, env sequences is able to rescue the defective retrovirus vector from macrophages, allowing gene transfer into muscle precursor cells. After fusion, these cells gave rise to genetically modified myotubes in vitro.
Collapse
Affiliation(s)
- E Parrish
- Généthon - CNRS URA 1922, 1 bis Rue de l'Internationale, 91002, Evry, France
| | | | | | | | | |
Collapse
|
11
|
Wang X, Smith GM, Xu XM. Preferential and bidirectional labeling of the rubrospinal tract with adenovirus-GFP for monitoring normal and injured axons. J Neurotrauma 2011; 28:635-47. [PMID: 21299337 DOI: 10.1089/neu.2010.1566] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The rodent rubrospinal tract (RST) has been studied extensively to investigate regeneration and remodeling of central nervous system (CNS) axons. Currently no retrograde tracers can specifically label rubrospinal axons and neurons (RSNs). The RST can be anterogradely labeled by injecting tracers into the red nucleus (RN), but accurately locating the RN is a technical challenge. Here we developed a recombinant adenovirus carrying a green fluorescent protein reporter gene (Adv-GFP) which can preferentially, intensely, and bi-directionally label the RST. When Adv-GFP was injected into the second lumbar spinal cord, the GFP was specifically transported throughout the entire RST, with peak labeling seen at 2 weeks post-injection. When Adv-GFP was injected directly into the RN, GFP was anterogradely transported throughout the RST. Following spinal cord injury (SCI), injection of Adv-GFP resulted in visualization of GFP in transected, spared, or sprouted RST axons bi-directionally. Thus Adv-GFP could be used as a novel tool for monitoring and evaluating strategies designed to maximize RST axonal regeneration and remodeling following SCI.
Collapse
Affiliation(s)
- Xiaofei Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
12
|
Abstract
A milestone of molecular medicine is the identification of dystrophin gene mutation as the cause of Duchenne muscular dystrophy (DMD). Over the last 2 decades, major advances in dystrophin biology and gene delivery technology have created an opportunity to treat DMD with gene therapy. Remarkable success has been achieved in treating dystrophic mice. Several gene therapy strategies, including plasmid transfer, exon skipping, and adeno-associated virus-mediated microdystrophin therapy, have entered clinical trials. However, therapeutic benefit has not been realized in DMD patients. Bridging the gap between mice and humans is no doubt the most pressing issue facing DMD gene therapy now. In contrast to mice, dystrophin-deficient dogs are genetically and phenotypically similar to human patients. Preliminary gene therapy studies in the canine model may offer critical insights that cannot be obtained from murine studies. It is clear that the canine DMD model may represent an important link between mice and humans. Unfortunately, our current knowledge of dystrophic dogs is limited, and the full picture of disease progression remains to be clearly defined. We also lack rigorous outcome measures (such as in situ force measurement) to monitor therapeutic efficacy in dystrophic dogs. Undoubtedly, maintaining a dystrophic dog colony is technically demanding, and the cost of dog studies cannot be underestimated. A carefully coordinated effort from the entire DMD community is needed to make the best use of the precious dog resource. Successful DMD gene therapy may depend on valid translational studies in dystrophin-deficient dogs.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
13
|
Wang D, Liu S, Mao Q, Zhao J, Xia H. A novel vector for a rapid generation of fiber-mutant adenovirus based on one step ligation and quick screening of positive clones. J Biotechnol 2011; 152:72-6. [DOI: 10.1016/j.jbiotec.2011.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/26/2011] [Accepted: 02/05/2011] [Indexed: 10/18/2022]
|
14
|
Liu L, Wang S, Shan B, Sang M, Liu S, Wang G. Advances in viral-vector systemic cytokine gene therapy against cancer. Vaccine 2010; 28:3883-7. [PMID: 20371389 DOI: 10.1016/j.vaccine.2010.03.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 03/21/2010] [Indexed: 01/31/2023]
Abstract
Current strategies for cancer gene therapy consist mainly of direct inhibition of tumor cell growth and activation of systemic host defense mechanisms. Cytokine gene-transduced tumor cells have been used as vaccines in clinical trials, which have shown good safety profiles and some local responses but substantial lack of systemic efficacy. Cytokines should be directed at the level of gene selection and delivery, in order to identify the optimal cytokine and achieve efficient and durable cytokine expression at the level of improving immune stimulation. In this review, we will summarize the current achievements of cytokine gene therapy, especially viral-vector, and their applications in cancer treatment. Additionally, we will also discuss and propose future perspectives about cancer gene therapy.
Collapse
Affiliation(s)
- Lihua Liu
- Research Center, the Fourth Clinical Hospital of Hebei Medical University and Hebei Cancer Institute, 12 Jiankanglu, Shijiazhuang, 050011, China
| | | | | | | | | | | |
Collapse
|
15
|
miR-122a-regulated expression of a suicide gene prevents hepatotoxicity without altering antitumor effects in suicide gene therapy. Mol Ther 2008; 16:1719-1726. [PMID: 18665157 DOI: 10.1038/mt.2008.159] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 07/02/2008] [Indexed: 01/11/2023] Open
Abstract
The combined use of adenovirus (Ad) vectors expressing herpes simplex virus thymidine kinase (HSVtk) and ganciclovir (GCV) offers a potential therapeutic strategy against cancer. However, intratumorally injected Ad vectors are disseminated into the systemic circulation and efficiently transduce the liver, resulting in severe hepatotoxicity. In order to overcome this problem, an Ad vector carrying a microRNA (miRNA)-regulated expression system was developed by inserting into the 3'-untranslated region (3'-UTR) of the expression cassette four tandem copies of sequences with perfect complementarity to miR-122a, which exhibits liver-specific expression. Transgene expression from the Ad vector carrying the miR-122a target sequences was 7- to 70-fold lower in cells with high miR-122a expression as compared to expression from a conventional Ad vector. Intratumoral injection of the Ad vector containing the miR-122a target sequences resulted in a 130- to 1,500-fold reduction in hepatic transgene products (without affecting the transgene expression in the tumor) when compared with those from a conventional Ad vector. In suicide gene therapy, the inclusion of the miR-122a target sequences in the HSVtk expression cassette achieved not only significant antitumor effects, but also a dramatic reduction in HSVtk/GCV-induced hepatotoxicity. These results indicate that Ad vectors that mediate miR-122a-regulated HSVtk expression provide a safe and efficient suicide gene therapy strategy.
Collapse
|
16
|
Kachi S, Esumi N, Zack DJ, Campochiaro PA. Sustained expression after nonviral ocular gene transfer using mammalian promoters. Gene Ther 2006; 13:798-804. [PMID: 16467860 DOI: 10.1038/sj.gt.3302700] [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: 02/08/2023]
Abstract
The CMV promoter drives high transgene expression and is one of the most commonly used promoters for gene transfer. Tissue-specific mammalian promoters provide an alternative, and it would be useful to have a system to directly compare them to viral promoters free from potential confounding vector-related effects. In this study, we describe how electroporation after subretinal injection of plasmid DNA can be used to perform comparative quantitative analysis of promoter activities. Luciferase assay of eyecup homogenates was carried out after coinjection/electroporation of pGL2, a plasmid containing the promoter fragment of interest coupled to the firefly luciferase gene, and pRL-CMV, a plasmid containing the CMV promoter coupled to the Renilla luciferase gene for normalization. This technique was used to compare activity of different fragments of the 5'-upstream region of the vitelliform macular dystrophy 2 (VMD2) gene, which is selectively expressed in the retinal pigmented epithelial (RPE) cells, and results indicated positive regulatory elements between -104 and -154 bp and between -424 and -585 bp. Addition of a fragment from intron 1 reduced the activity of the -585/+38 bp fragment by 75%. Deletion analysis implicated a 342 bp region near the 5'-end of intron 1 in the repression. Results of transient transfections in two cell lines that constitutively express VMD2 were similar, and results in transgenic mice were consistent, providing validation for promoter analysis by in vivo electroporation. We then explored the time course of expression of the -585/+38 VMD2 promoter fragment and found that compared to cassettes driven by CMV or SV40 promoters, which showed peak luciferase activity on day 2 followed by a rapid decrease in activity, the VMD2 promoter fragment showed lower activity initially, but the activity was sustained for up to 56 days (longest time point measured). A promoter fragment from another RPE-specific gene, Rpe65, showed a similar pattern of sustained expression for at least 112 days. These data indicate that nonviral gene transfer can be used to quantitatively evaluate the activity of promoter fragments independent of influence from viral vectors. A potentially important finding using this new technique is the demonstration that relatively sustained passenger gene expression can be achieved with nonviral gene transfer using mammalian rather than viral promoters.
Collapse
Affiliation(s)
- S Kachi
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
17
|
Man LX, Park JC, Terry MJ, Mason JM, Burrell WA, Liu F, Kimball BY, Moorji SM, Lee JA, Breitbart AS. Lentiviral gene therapy with platelet-derived growth factor B sustains accelerated healing of diabetic wounds over time. Ann Plast Surg 2006; 55:81-6; discussion 86. [PMID: 15985796 DOI: 10.1097/01.sap.0000168211.76318.fa] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The treatment of diabetic wounds is a formidable clinical challenge. In this study, lentiviral vectors carrying the human platelet-derived growth factor B (PDGF-B) gene were used to treated diabetic mouse wounds. Full-thickness 2.0-cm x 2.0-cm excisional wounds were created on the dorsa of genetically diabetic C57BL/KsJ-m+/+Lepr(db) mice. Lentiviral vectors containing the PDGF-B gene were injected into the wound margins and base. Mice were killed at 14-, 21-, and 35-day intervals. Measurement of the residual epithelial gap showed a trend towards increased healing in lentiviral PDGF-treated wounds compared with untreated and saline-treated wounds at all time points. At 21 days, there was significantly increased healing in lentiviral PDGF-treated wounds (0.98+/-0.17 cm) compared with saline-treated wounds (1.22+/-0.30 cm; P<0.05). Immunohistochemistry for CD31 revealed significantly increased neovascularization in lentiviral PDGF-treated wounds compared with untreated and saline-treated wounds at 14 and 21 days (P<0.01). Picrosirius red staining demonstrated thicker and more highly organized collagen fibers in treated wounds compared with untreated and saline-treated wounds. Quantitative analysis of collagen content showed a 3.5-fold and 2.3-fold increase in lentiviral PDGF-treated wounds versus untreated and saline-treated wounds, respectively (P<0.01). Lentiviral gene therapy with PDGF-B can sustain diabetic wound healing over time and may possess promising potential in the clinical setting.
Collapse
Affiliation(s)
- Li-Xing Man
- Division of Plastic and Reconstructive Surgery, New York Presbyterian Hospital, Columbia-Presbyterian Medical Center, Columbia University College of Physicians and Surgeons, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Tomioka R, Rockland KS. Improved Golgi-like Visualization in Retrogradely Projecting Neurons after EGFP-Adenovirus Infection in Adult Rat and Monkey. J Histochem Cytochem 2006; 54:539-48. [PMID: 16344324 DOI: 10.1369/jhc.5a6838.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An adenovirus vector was generated using a neuron-specific promoter synapsin I and enhanced green fluorescent protein (EGFP) reporter (AdSynEGFP). In addition, two modifications were identified that resulted in robust and reliable retrograde transport and EGFP expression after injection of the virus into three different brain regions in adult rats (medial prefrontal cortex, posterior thalamic nuclear group, and CA1). These are post-injection survival times of 14 days and addition of high concentrations of NaCl (≥600 mM) to the injection buffer. These modifications resulted in obvious improvement in the intensity of the EGFP signal and in the number of labeled cells. Use of anti-EGFP in immunofluorescence or immunoperoxidase processing further enhanced the signal so that Golgi-like filling of dendritic spines and axon collaterals was routinely achieved. Effectiveness of the AdSynEGFP for Golgi-like filling was confirmed in one rhesus monkey with injections in visual area V4. Because of the long-term viability of the infected neurons (at least up to 28 days in rats and 22 days in monkey), this AdSynEGFP is suitable for use in microcircuitry studies in combination with other fluorescently tagged elements, including anterogradely labeled extrinsic projections. The native EGFP signal (without antibody enhancement) may be sufficient for studies involving cultured cells or slices. (J Histochem Cytochem 54:539-548, 2006)
Collapse
Affiliation(s)
- Ryohei Tomioka
- Laboratory for Cortical Organization and Systematics, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
| | | |
Collapse
|
19
|
Lee JA, Conejero JA, Mason JM, Parrett BM, Wear-Maggitti KD, Grant RT, Breitbart AS. Lentiviral Transfection with the PDGF-B Gene Improves Diabetic Wound Healing. Plast Reconstr Surg 2005; 116:532-8. [PMID: 16079687 DOI: 10.1097/01.prs.0000172892.78964.49] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The treatment of diabetic wounds remains a difficult challenge. The present study investigates whether platelet-derived growth factor (PDGF) lentiviral gene therapy can improve diabetic wound healing in the diabetic db/dbmouse. METHODS PDGF cDNA was cloned and lentiviral vectors were constructed with either the PDGF-B or green fluorescence protein (GFP) gene. A 2 x 2-cm full-thickness dermal wound was made on each db/db mouse. Animals were divided into three groups, with eight animals in each group as follows: group I, empty wound; group II, lentiviral PDGF; and group III, lentiviral GFP. Lentiviral vectors were injected into the wounds and healing was assessed at 21 days. Harvested wounds were assessed for residual epithelial gap, granulation tissue area, PDGF expression, collagen formation (picrosirius red), and angiogenesis (CD31 staining). RESULTS Lentiviral vectors were constructed and transfected dermal fibroblasts demonstrated in vitro production of PDGF mRNA as measured by reverse-transcriptase polymerase chain reaction. Immunohistochemistry for PDGF confirmed successful in vivo transfection of the PDGF gene. At 21 days, reepithelialization and granulation tissue area were similar in all groups. However, there was a statistically significant increase in angiogenesis and substantially thicker, more coherently aligned collagen fibers in the PDGF group compared with controls. CONCLUSIONS PDGF lentiviral vectors were successfully transfected into the regenerated dermis in diabetic wounds. Although reepithelialization was similar among the groups, there was enhanced angiogenesis and collagen deposition in the lentiviral PDGF group. These results demonstrate that lentiviral PDGF transfection of the diabetic wound enhances PDGF production, improves vascularization and collagen organization, and has potential clinical applications in diabetic wound treatment.
Collapse
Affiliation(s)
- James A Lee
- Division of Plastic and Reconstructive Surgery, New York Presbyterian Hospital, Columbia University Medical Center, College of Physicians and Surgeons, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Hoffmann D, Jogler C, Wildner O. Effects of the Ad5 upstream E1 region and gene products on heterologous promoters. J Gene Med 2005; 7:1356-66. [PMID: 15945123 DOI: 10.1002/jgm.771] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND All recombinant adenovirus vectors contain the upstream region of the E1A gene comprising the viral origin of replication, encapsidation signal, and cis-acting regulatory elements for transcription of the E1A and other early genes. Using different reporter genes, some previous studies demonstrated the maintenance of heterologous promoter specificity in the adenoviral context, while others reported that adenoviral sequences interfere with promoter activity. METHODS Plasmid DNA-based luciferase reporter gene assays and adenovirus type 5 (Ad5) infection were combined to examine the effect of the Ad5 (nt 1-353) element and/or adenoviral gene products on tissue-specific (Midkine (MK) and COX-2), cell cycle associated (Ki-67 and E2F1) and viral promoters (Ad5 E1, Ad5 E4 and SV40). As a proof of concept, data were verified in the setting of recombinant replication-defective and replication-competent adenoviral vectors. RESULTS Viral and E2F1 promoter activities were enhanced by the Ad5 (nt 1-353) segment by approximately 100% and 145%, respectively, regardless of its position. A polyadenylation sequence (polyA) upstream of the promoter had no effect, confirming an enhancer element within the Ad5 (nt 1-353) segment. Ad5 (nt 1-353) increased COX-2 promoter activity by 146% but was blocked by an upstream polyA, indicating a cryptic transcription start site. When placing the reporter gene cassette in a replication-defective adenovirus, similar data were obtained. In the plasmid vector-based system, adenoviral gene products transactivated the E2F1 and viral promoters by 194%, 19%, 67%, and 16%, respectively. Tissue-specific promoter activities were not significantly affected by the Ad5 (nt 1-353) segment, nor adenoviral gene products. In concert with these data, we were able to target replication-competent adenoviral vectors with the COX-2 promoter, but not with the cell cycle associated promotor. CONCLUSIONS The adenovirus E1A upstream regulatory region and gene products interact with some but not all heterologous promoters. Often, the basal promoter activity can be reduced with an upstream polyA. Since the data obtained in our plasmid vector-based assay with internal control and infection with adenovirus could be confirmed in the adenoviral setting, our system might be suitable to speed up the identification of promoters which maintain their specificity in the adenoviral context and circumvent the problems associated with determining infectious adenovirus titers.
Collapse
Affiliation(s)
- Dennis Hoffmann
- Ruhr-Universität Bochum, Abteilung für Molekulare und Medizinische Virologie, Bldg. MA, Rm. 6/40, D-44801 Bochum, Germany
| | | | | |
Collapse
|
21
|
Watanabe T, Okuda Y, Nonoguchi N, Zhao MZ, Kajimoto Y, Furutama D, Yukawa H, Shibata MA, Otsuki Y, Kuroiwa T, Miyatake SI. Postischemic intraventricular administration of FGF-2 expressing adenoviral vectors improves neurologic outcome and reduces infarct volume after transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2004; 24:1205-13. [PMID: 15545913 DOI: 10.1097/01.wcb.0000136525.75839.41] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Fibroblast growth factor (FGF)-2 is a potent neurotrophic and angiogenic peptide. To examine possible protective effects of FGF-2 gene expression against transient focal cerebral ischemia in rats, a replication defective, recombinant adenovirus vector expressing FGF-2, was injected intraventricularly 2 hours after middle cerebral artery occlusion (MCAO). The treatment group showed significant recovery compared with the vehicle-treated groups in terms of serial neurologic severity scores over the 35 days after MCAO. Further, 2,3,5-triphenyltetrazolium chloride staining showed that FGF-2 gene transfer decreased infarct volume by 44% as compared with that in the vehicle-treated groups at 2 days after MCAO. The same tendency of gene transfer effects on infarct volume was confirmed at 35 days after MCAO with hematoxylin/eosin staining. Enzyme-linked immunosorbent assay revealed that FGF-2 concentration was increased significantly at 2 days after MCAO, not only in cerebrospinal fluid but also in cerebral substance in the lesioned and treated animals. These results suggested that FGF-2 gene transfer using these adenoviral vectors might be a useful modality for the treatment of occlusive cerebrovascular disease even after the onset of stroke.
Collapse
Affiliation(s)
- Takuji Watanabe
- Department of Neurosurgery, Osaka Medical College, Takatsuki 569-8686, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lin ZH, Fukuda N, Suzuki R, Takagi H, Ikeda Y, Saito S, Matsumoto K, Kanmatsuse K, Mugishima H. Adenovirus-Encoded Hammerhead Ribozyme to PDGF A-Chain mRNA Inhibits Neointima Formation after Arterial Injury. J Vasc Res 2004; 41:305-13. [PMID: 15192266 DOI: 10.1159/000078928] [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] [Received: 11/21/2003] [Accepted: 04/05/2004] [Indexed: 11/19/2022] Open
Abstract
To develop a strategy for gene therapy of restenosis following coronary angioplasty, we examined the effects of a recombinant adenovirus vector encoding a hammerhead ribozyme specific for rat platelet-derived growth factor (PDGF) A-chain mRNA (Ad. Ribozyme) and a control recombinant adenovirus vector encoding the Escherichia coli LacZ gene (Ad.LacZ) on neointima formation in rat carotid artery after balloon injury. Ad. Ribozyme (10(8) PFU/ml) markedly reduced the increased expression of PDGF A-chain mRNA and protein. Ad. Ribozyme significantly decreased the intima/media ratio (68%) of the injured artery, whereas Ad.LacZ had no effect on the intima/media ratio. Most carotid arteries developed thrombi by 14 days after balloon injury, whereas Ad. Ribozyme completely inhibited thrombus formation. Expression of thromboxane A2 (TXA2) receptor mRNA was significantly increased after balloon injury. Ad. Ribozyme significantly decreased the levels of TXA2 receptor. Expression of prostaglandin I2 (PGI2) synthase mRNA was significantly decreased after balloon injury. Ad. Ribozyme significantly increased levels of PGI2 synthase mRNA after balloon injury. The observation that adenovirus-encoded ribozyme to PDGF A-chain inhibits neointima formation may serve as a novel strategy to prevent restenosis after coronary angioplasty. Inhibition of growth factors by genetic approaches may yield new insights into the mechanisms underlying responses to vascular injury and lead to new therapeutic applications.
Collapse
Affiliation(s)
- Zhi-Hong Lin
- Second Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The main objective in gene therapy is the development of efficient, non-toxic gene carriers that can encapsulate and deliver foreign genetic materials into specific cell types such as cancerous cells. During the past two decades, enormous research in the area of gene delivery has been conducted worldwide, in particular for cancer gene therapy application. Viral vectors are biological systems derived from naturally evolved viruses capable of transferring their genetic materials into the host cells. Many viruses including retrovirus, adenovirus, herpes simplex virus (HSV), adeno-associated virus (AAV) and pox virus have been modified to eliminate their toxicity and maintain their high gene transfer capability. The limitations associated with viral vectors, however, in terms of their safety, particularly immunogenicity, and in terms of their limited capacity of transgenic materials, have encouraged researchers to increasingly focus on non-viral vectors as an alternative to viral vectors. Non-viral vectors are generally cationic in nature. They include cationic polymers such as poly(ethylenimine) (PEI) and poly(L-lysine) (PLL), cationic peptides and cationic liposomes. The newly described liposomal preparation LPD (liposomes/protamine/DNA), for example, has shown superiority over conventional liposomes/DNA complexes (lipoplexes). Although non-viral vectors are less efficient than viral ones, they have the advantages of safety, simplicity of preparation and high gene encapsulation capability. This article reviews the most recent studies highlighting the advantages and the limitations of various types of gene delivery systems used in cancer gene therapy.
Collapse
Affiliation(s)
- Anas El-Aneed
- School of Pharmacy, Memorial University of Newfoundland, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6.
| |
Collapse
|
24
|
Oshikawa J, Otsu K, Toya Y, Tsunematsu T, Hankins R, Kawabe JI, Minamisawa S, Umemura S, Hagiwara Y, Ishikawa Y. Insulin resistance in skeletal muscles of caveolin-3-null mice. Proc Natl Acad Sci U S A 2004; 101:12670-5. [PMID: 15314230 PMCID: PMC515114 DOI: 10.1073/pnas.0402053101] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes is preceded by the development of insulin resistance, in which the action of insulin is impaired, largely in skeletal muscles. Caveolin-3 (Cav3) is a muscle-specific subtype of caveolin, an example of a scaffolding protein found within membranes. Cav is also known as growth signal inhibitor, although it was recently demonstrated that the genetic disruption of Cav3 did not augment growth in mice. We found, however, that the lack of Cav3 led to the development of insulin resistance, as exemplified by decreased glucose uptake in skeletal muscles, impaired glucose tolerance test performance, and increases in serum lipids. Such impairments were markedly augmented in the presence of streptozotocin, a pancreatic beta cell toxin, suggesting that the mice were susceptible to severe diabetes in the presence of an additional risk factor. Insulin-stimulated activation of insulin receptors and downstream molecules, such as IRS-1 and Akt, was attenuated in the skeletal muscles of Cav3 null mice, but not in the liver, without affecting protein expression or subcellular localization. Genetic transfer of Cav3 by needle injection restored insulin signaling in skeletal muscles. Our findings suggest that Cav3 is an enhancer of insulin signaling in skeletal muscles but does not act as a scaffolding molecule for insulin receptors.
Collapse
Affiliation(s)
- Jin Oshikawa
- Departments of Physiology and Medicine, Yokohama City University School of Medicine, 3-9 Fukuura Kanazawa, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yang HS, Lee H, Kim SJ, Lee WW, Yang YJ, Moon DH, Park SW. Imaging of human sodium-iodide symporter gene expression mediated by recombinant adenovirus in skeletal muscle of living rats. Eur J Nucl Med Mol Imaging 2004; 31:1304-11. [PMID: 15221289 DOI: 10.1007/s00259-004-1570-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE We evaluated the feasibility of non-invasive imaging of recombinant adenovirus-mediated human sodium-iodide symporter (hNIS) gene expression by (99m)TcO(4)(-) scintigraphy in skeletal muscle of rats. METHODS Replication-defective recombinant adenovirus encoding hNIS gene [Rad-CMV-hNIS 5x10(7), 2x10(8) or 1x10(9) plaque forming units (pfu)] or beta-galactosidase gene (Rad-CMV-LacZ 1x10(9) pfu) was injected into the right biceps femoris muscle of rats ( n=5-6 for each group). Three days after gene transfer, scintigraphy was performed using a gamma camera 30 min after injection of (99m)TcO(4)(-) (1.85 MBq). An additional two rats injected with 1x10(9) pfu of Rad-CMV-hNIS underwent (99m)TcO(4)(-) scintigraphy with sodium perchlorate. After the imaging studies, rats were sacrificed for assessment of the biodistribution of (99m)TcO(4)(-) and measurement of hNIS mRNA expression. RESULTS In all the rats injected with 1x10(9) pfu of Rad-CMV-hNIS, hNIS expression was successfully imaged by (99m)TcO(4)(-) scintigraphy, while rats injected with Rad-CMV-LacZ or lower doses of Rad-CMV-hNIS failed to show uptake. The biodistribution studies indicated that a significantly different amount of (99m)TcO(4)(-) was retained in the liver ( p<0.001) and the right muscle ( p<0.05), with the highest uptake in rats injected with 1x10(9) pfu of Rad-CMV-hNIS. The muscular hNIS mRNA level quantified by real-time reverse transcription-polymerase chain reaction was significantly higher in rats injected with 1x10(9) pfu of Rad-CMV-hNIS ( p<0.05), with a positive correlation with the imaging counts ( r=0.810, p<0.05) and the biodistribution ( r=0.847, p<0.001). Hot spots in rats injected with 1x10(9) pfu of Rad-CMV-hNIS were specifically inhibited by sodium perchlorate. CONCLUSION This study illustrated that (99m)TcO(4)(-) scintigraphy can monitor Rad-CMV-hNIS-mediated gene expression in skeletal muscle of rats, non-invasively and quantitatively.
Collapse
Affiliation(s)
- Hyun Suk Yang
- Department of Internal Medicine (Cardiology), Asan Medical Center, University of Ulsan College of Medicine, 388-1 Pungnap-dong, Songpa-gu, 138-736 Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
26
|
Mercier S, Verhaagh S, Goudsmit J, Lemckert A, Monteil M, Havenga M, Eloit M. Adenovirus fibre exchange alters cell tropism in vitro but not transgene-specific T CD8+ immune responses in vivo. J Gen Virol 2004; 85:1227-1236. [PMID: 15105539 DOI: 10.1099/vir.0.79846-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Gene transfer with recombinant adenoviruses (rAds) is a powerful means of inducing an immune response against a transgene product. However, little is known about the mechanisms that underlie the induction of the immune response after intramuscular inoculation of adenovirus and, in particular, the relative role of the different cell types transduced. Several studies have suggested that CD8+ cytotoxic T lymphocyte responses elicited after inoculation of adenoviruses (Ads) are induced both by direct transduction of antigen presenting cells (APCs) and by cross-priming. In the present study, a library of fibre-chimeric rAds was screened in order to identify rAds with distinct capacities to express transgene product in murine cell types naturally found in muscle, i.e. myoblasts, endothelial cells (both representing non-APCs) and dendritic cells (representing APCs). Four selected pseudotypes, differing in their ability to infect muscular cells were used to immunize C57BL/6 mice. The relationship between the capacity to transduce non-APC or APC in vitro and the ability to induce humoral and cellular responses against the beta-galactosidase antigen after intramuscular inoculation were studied. Results indicate that CD8+ T cell responses against the beta-galactosidase antigen were similar after inoculation of the four viruses, thus revealing no direct relationship with their ability to transduce myoblasts, endothelial cells or dendritic cells in vitro.
Collapse
Affiliation(s)
- S Mercier
- UMR INRA-AFSSA-ENVA 1161, Virologie, Ecole Nationale Vétérinaire d'Alfort, 94704 Maisons-Alfort Cedex, France
| | - S Verhaagh
- Crucell Holland BV, PO Box 2048, 2301 CA Leiden, The Netherlands
| | - J Goudsmit
- Crucell Holland BV, PO Box 2048, 2301 CA Leiden, The Netherlands
| | - A Lemckert
- Crucell Holland BV, PO Box 2048, 2301 CA Leiden, The Netherlands
| | - M Monteil
- UMR INRA-AFSSA-ENVA 1161, Virologie, Ecole Nationale Vétérinaire d'Alfort, 94704 Maisons-Alfort Cedex, France
| | - M Havenga
- Crucell Holland BV, PO Box 2048, 2301 CA Leiden, The Netherlands
| | - M Eloit
- UMR INRA-AFSSA-ENVA 1161, Virologie, Ecole Nationale Vétérinaire d'Alfort, 94704 Maisons-Alfort Cedex, France
| |
Collapse
|
27
|
Breitbart AS, Laser J, Parrett B, Porti D, Grant RT, Grande DA, Mason JM. Accelerated diabetic wound healing using cultured dermal fibroblasts retrovirally transduced with the platelet-derived growth factor B gene. Ann Plast Surg 2004; 51:409-14. [PMID: 14520070 DOI: 10.1097/01.sap.0000084461.83554.71] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The treatment of diabetic wounds is a considerable clinical challenge. In this study, mouse dermal fibroblasts retrovirally transduced with the human platelet-derived growth factor B (PDGF-B) gene were used to treat diabetic mouse wounds. The PDGF-B gene was obtained from human umbilical vein endothelial cells, cloned into retroviral vectors, and introduced into diabetic mouse C57B1/ks-db/db dermal fibroblasts. In vitro results demonstrated production of PDGF-B protein by these transduced cells at steady-state levels of 1000 ng PDGF-B/10(6) cells/24 hours, and expression of PDGF-B mRNA. These cells were seeded onto polyglycolic acid scaffold matrices and used to treat diabetic mouse 20-mm x 20-mm full-thickness excisional dorsal skin wounds. Measurement of the residual epithelial gap at 21 days showed significantly accelerated healing (P < 0.05) of wounds treated with PDGF-transduced cells (epithelial gap 10.46 +/- 1.20 mm) compared with untreated wounds (14.66 +/- 0.591 mm), wounds treated with polyglycolic acid alone (14.80 +/- 0.575 mm), or wounds treated with negative control LNCX-transduced cells (13.76 +/- 0.831 mm). Immunohistochemical staining showed intense staining for PDGF in wounds treated with PDGF-B-transduced cells. This study demonstrates the promising potential for gene therapy in diabetic wound healing.
Collapse
Affiliation(s)
- Arnold S Breitbart
- Division of Plastic and Reconstructive Surgery, New York Presbyterian Hospital, Columbia-Presbyterian Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Saulnier P, Vidaud M, Gautier E, Motté N, Bellet D, Escudier B, Wilson D, Yver A. Development and validation of a real-time PCR assay for the detection and quantitation of p53 recombinant adenovirus in clinical samples from patients treated with Ad5CMV-p53 (INGN 201). J Virol Methods 2003; 114:55-64. [PMID: 14599679 DOI: 10.1016/j.jviromet.2003.08.012] [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: 11/17/2022]
Abstract
The purpose of this study was to assess the usefulness of real-time PCR as a quantitative, highly reproducible, and sensitive method, for detecting and quantifying p53 recombinant adenovirus in biological samples from cancer patients receiving injections of Ad5CMV-p53. The dynamic range of this real-time PCR-based assay was wide (at least five orders of magnitude). Our assay used an internal positive control in the same PCR tube that is capable of detecting residual PCR inhibitors. Serial spiked samples in plasma with known quantities of Ad5CMV-p53 were evaluated. The minimum detection limit was 2 pfu per PCR (approximately 50 pfu per ml of plasma) and the quantification values were reproducible. A total of 2069 controls tested with 1780 plasma samples from 286 patients enrolled in gene therapy trials using Ad5CMV-p53 were investigated. Using calibrators to adjust the quantitation value, the results confirmed the good performance of the assay. In conclusion, the high sensitivity, simplicity and reproducibility of the real-time Ad5CMV-p53 assay, allowing screening of large numbers of samples, combined with its wide dynamic range, make this method particularly suitable for monitoring gene therapy trials.
Collapse
Affiliation(s)
- Patrick Saulnier
- Centre de Référence IGR/Aventis Gencell, Institut Gustave-Roussy, 39, rue Camille Desmoulins, F-94805 Villejuif, France.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Chuang IC, Jhao CM, Yang CH, Chang HC, Wang CW, Lu CY, Chang YJ, Lin SH, Huang PL, Yang LC. Intramuscular electroporation with the pro-opiomelanocortin gene in rat adjuvant arthritis. Arthritis Res Ther 2003; 6:R7-R14. [PMID: 14979933 PMCID: PMC400409 DOI: 10.1186/ar1014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2003] [Revised: 09/18/2003] [Accepted: 09/30/2003] [Indexed: 11/10/2022] Open
Abstract
Endogenous opioid peptides have an essential role in the intrinsic modulation and control of inflammatory pain, which could be therapeutically useful. In this study, we established a muscular electroporation method for the gene transfer of pro-opiomelanocortin (POMC) in vivo and investigated its effect on inflammatory pain in a rat model of rheumatoid arthritis. The gene encoding human POMC was inserted into a modified pCMV plasmid, and 0-200 microg of the plasmid-POMC DNA construct was transferred into the tibialis anterior muscle of rats treated with complete Freund's adjuvant (CFA) with or without POMC gene transfer by the electroporation method. The safety and efficiency of the gene transfer was assessed with the following parameters: thermal hyperalgesia, serum adrenocorticotropic hormone (ACTH) and endorphin levels, paw swelling and muscle endorphin levels at 1, 2 and 3 weeks after electroporation. Serum ACTH and endorphin levels of the group into which the gene encoding POMC had been transferred were increased to about 13-14-fold those of the normal control. These levels peaked 1 week after electroporation and significantly decreased 2 weeks after electroporation. Rats that had received the gene encoding POMC had less thermal hypersensitivity and paw swelling than the non-gene-transferred group at days 3, 5 and 7 after injection with CFA. Our promising results showed that transfer of the gene encoding POMC by electroporation is a new and effective method for its expression in vivo, and the analgesic effects of POMC cDNA with electroporation in a rat model of rheumatoid arthritis are reversed by naloxone.
Collapse
Affiliation(s)
- I-Chuan Chuang
- Gene Therapy Laboratory, Tajen Institute of Technology, Pingtung, 907, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kubo S, Mitani K. A new hybrid system capable of efficient lentiviral vector production and stable gene transfer mediated by a single helper-dependent adenoviral vector. J Virol 2003; 77:2964-71. [PMID: 12584321 PMCID: PMC149763 DOI: 10.1128/jvi.77.5.2964-2971.2003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2002] [Accepted: 12/03/2002] [Indexed: 11/20/2022] Open
Abstract
To achieve efficient and sustained gene expression, we developed a new lentivirus/adenovirus hybrid vector (LA vector) that encodes sequences required for production of a human immunodeficiency virus-based lentiviral vector (i.e., a lentiviral vector, a gag/pol/rev expression cassette, a tetracycline-inducible envelope cassette, and the tetracycline-inducible transcriptional activator cassette) in a single helper-dependent adenovirus vector backbone. Via either transfection or infection, human cell lines transduced with the LA vector produced a lentiviral vector in a doxycycline-dependent manner at titers up to 10(5) to 10(6) green fluorescent protein transducing units per ml, which are comparable to the titers obtained by conventional multiple plasmid transfection methods. Efficient spread and persistent expression of the transgene were observed in cells maintained in long-term culture that had been infected with the LA vector. Furthermore, when cocultured with adherent cells infected with the LA vector, the human T-cell leukemia cell line was successfully transduced with a marker gene. This LA vector possesses the advantages of efficient gene transfer from an adenoviral vector and stable integration from a lentiviral vector; therefore, it might have potential for a variety of gene therapy applications.
Collapse
Affiliation(s)
- Shuji Kubo
- Department of Microbiology, Immunology and Molecular Genetics, UCLA School of Medicine, Los Angeles, Los Angeles, California 90095-1747, USA
| | | |
Collapse
|
31
|
Lowenstein PR, Suwelack D, Hu J, Yuan X, Jimenez-Dalmaroni M, Goverdhana S, Castro MG. Nonneurotropic adenovirus: a vector for gene transfer to the brain and gene therapy of neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 55:3-64. [PMID: 12968530 PMCID: PMC2902245 DOI: 10.1016/s0074-7742(03)01001-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Pedro R Lowenstein
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Department of Medicine, UCLA, Los Angeles, California 90048, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Dickson G, Roberts ML, Wells DJ, Fabb SA. Recombinant micro-genes and dystrophin viral vectors. Neuromuscul Disord 2002; 12 Suppl 1:S40-4. [PMID: 12206793 DOI: 10.1016/s0960-8966(02)00080-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An effective gene therapy for Duchenne muscular dystrophy ideally relies on the ability to provide long-term expression to muscle tissue of the missing protein, dystrophin. Early work in the mdx mouse using a 6.3 kb mini-dystrophin cDNA, carried out in either adenoviral or retroviral vectors was generally successful, however, expression was only transient. In an attempt to remedy this problem, two approaches are being investigated. The first of these is a hybrid vector system that combines the efficacy of gene transfer into skeletal muscle of adenoviral vectors with the long-term stability of retroviral vectors. The second utilises the inherently efficient transducing properties and stability of the adeno-associated viral delivery system. Using highly truncated micro-dystrophin cDNAs we have shown that both vector systems were able to restore dystrophin and dystrophin-associated protein expression at the plasma membrane of mdx mice for prolonged periods of time. Additionally, evaluation of central nucleation indicated a significant inhibition of degenerative dystrophic muscle pathology. These studies suggest that hybrid adenoviral-retroviral and adeno-associated viral vectors are capable of ameliorating dystrophic pathology at the cellular level and as such are useful tools in the development of a gene therapy for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- G Dickson
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Surrey TW20 0EX, UK.
| | | | | | | |
Collapse
|
33
|
Pitard B, Pollard H, Agbulut O, Lambert O, Vilquin JT, Cherel Y, Abadie J, Samuel JL, Rigaud JL, Menoret S, Anegon I, Escande D. A nonionic amphiphile agent promotes gene delivery in vivo to skeletal and cardiac muscles. Hum Gene Ther 2002; 13:1767-75. [PMID: 12396628 DOI: 10.1089/104303402760293592] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Direct injection of naked DNA into skeletal or cardiac muscle induces detectable gene expression. Although this provides a practical system for transgene expression, the reported efficacy is too low to confer a therapeutic benefit. By following a rational strategy based on the supramolecular structures adopted by active complexes, we have discovered a novel nonionic amphiphile synthetic agent [poly(ethyleneoxide)(13)-poly(propyleneoxide)(30)-poly(ethyleneoxide)(13) block copolymer; PE6400] that enables gene expression in up to 35% of muscle fibers from mouse tibial cranial muscle. PE6400 abolishes the ceiling effect on transgene expression of increasing amounts of naked DNA and permits long-term expression of the beta-galactosidase reporter gene in immunologically tolerant transgenic rats. This improvement in gene expression over naked DNA was observed irrespective of the reporter gene, ranging from 0.7 to 3.4 kb, and of the animal model used. In skeletal muscle, the PE6400 formulation led to a level of transfection efficiency similar to that obtained by electrotransfer. PE6400 also promotes high transgene expression in cardiac muscle. In contrast, PE6400-DNA formulations were inefficient in vitro in established cell lines and in isolated cardiomyocytes. When microinjected into the cell cytoplasm, PE6400 promotes DNA trafficking into the nucleus and induces gene expression. PE6400 provides a simple gene delivery system for skeletal and myocardial gene transfer. We propose that the PE6400 formulation could serve for the treatment of diseases primarily affecting muscle or for the expression of therapeutic proteins for local or systemic benefit.
Collapse
Affiliation(s)
- Bruno Pitard
- Inserm U533, Faculté de Médecine, 44000 Nantes, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Zhou HS, Zhao T, Rao XM, Beaudet AL. Production of helper-dependent adenovirus vector relies on helper virus structure and complementing. J Gene Med 2002; 4:498-509. [PMID: 12221643 DOI: 10.1002/jgm.301] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The helper-dependent (HD) adenoviral (Ad) vector relies on a helper virus to provide viral proteins for vector amplification. HD-Ad vectors can significantly increase therapeutic gene expression and improve safety. However, the yield of an HD-Ad vector is generally lower than that of an E1-deleted first-generation vector, likely due to the alterations in viral E3 or packaging regions of a helper virus that attenuate its replication and complementing for an HD-Ad vector. METHODS To study this question and improve HD-Ad vector production, we have generated four different helper viruses with a wild-type or deleted E3 region, and with a relocated loxP. We have also constructed a first-generation vector with a wild-type E3 region and without the loxP site. We compared the replication of these viruses in Cre-positive and -negative cells and studied their complementing for HD-Ad vector production. RESULTS Viruses with deleted E3 formed smaller plaques and produced lower titer compared with viruses containing the E3 region. The site where a loxP is inserted can also affect virus replication. Higher yield of HD-Ad vector was obtained when a helper virus with wild-type E3 was used. We also showed that deletion of the packaging signal in a helper virus through loxP/Cre interaction decreased the viral DNA complementing ability. CONCLUSIONS Although the E3 region is not essential for adenovirus replication in vivo, deletion of this region attenuates virus replication. Production of HD-Ad vector can be further improved by modifications in helper virus structure.
Collapse
Affiliation(s)
- Heshan Sam Zhou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
36
|
Abstract
Transfer of therapeutic genes into muscle tissue has promise for the treatment of a variety of muscular dystrophies. Various vectors have been used to deliver genes to skeletal muscle but their application has faced several major limitations including: (1) the lack of transgene persistence caused by the immune rejection of transduced myofibers and/or vector toxicity, and (2) the maturation dependence of viral transduction. While the immunorejection and/or cytotoxic problems are being overcome with the development of new vectors, maturation-dependent viral transduction is still a major hurdle in gene transfer to skeletal muscle. Poor adenoviral transduction in mature myofibers has been attributed to: (1) the extracellular matrix of mature myofibers may form a physical barrier and prevent the passage of large viral particles; (2) viral receptors are down-regulated with muscle maturation; and (3) loss of myoblasts with muscle maturation, which serve as intermediaries in the viral transduction. In this review, we will focus on recent developments in overcoming those hurdles of gene therapy in skeletal muscle, especially to adenovirus (Ad), including: (1) new mutant vectors lacking all viral genes to decrease immunogenicity, and hence, improve persistence of transgene expression in muscle in vivo; (2) using tissue specific promoters to evade immunorejection; (3) permeabilization of the extracellular matrix; (4) modifying the viral receptors in mature myofibers; and (5) myoblast or muscle stem cell mediated ex vivo gene transfer.
Collapse
Affiliation(s)
- Baohong Cao
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
37
|
Abstract
Over the past decade, the unprecedented growth in science and technology has fueled the development of novel treatment strategies to combat disease. The creative and innovative efforts of scientists and clinicians to overcome the multitude of unforeseen obstacles to success is no better exemplified than in the field of cancer gene therapy. Since its inception, developers of cancer gene therapy have been charged with the challenge of altering basic tumor biology or, alternatively, the host responses for the purpose of tumor eradication and prevention. Several major therapeutic strategies have emerged from preclinical studies, and results from these early studies hold promise for altering the clinical outcome in a variety of malignancies. These strategies may be broadly subcategorized and range in intent from alteration of the tumor cell phenotype by replacement of defective cellular response genes (e.g., mutated or deleted tumor suppressor genes) to the enhancement of the immunological response to cancer (e.g., amplification of the cell surface antigen signature or modulation of the host response). Not surprisingly, the increasingly intricate nature of tumor biology revealed over the past several years has effectively raised the bar of success for those involved in the development of effective molecular and cancer gene therapy strategies. This, in turn, has led to the development of more complex therapies that frequently draw upon multiple disciplines in an effort to optimize treatment response.
Collapse
Affiliation(s)
- James C Cusack
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Cox Building, Room 626, 100 Blossom Street, Boston, MA 02114, USA.
| | | |
Collapse
|
38
|
Huang YZ, Wang Q, Won S, Luo ZG, Xiong WC, Mei L. Compartmentalized NRG signaling and PDZ domain-containing proteins in synapse structure and function. Int J Dev Neurosci 2002; 20:173-85. [PMID: 12175853 DOI: 10.1016/s0736-5748(02)00011-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The synapse-specific synthesis of the acetylcholine receptor (AChR) is mediated by multiple mechanisms including compartmentalized signaling induced by neuregulin (NRG). This paper presents evidence that NRG receptors--ErbB receptor tyrosine kinases interact with distinct PDZ domain-containing proteins that are localized at the neuromuscular junction (NMJ). ErbB4 associates with the PSD-95 (also known as SAP90)-family members including PSD-95, SAP97, and SAP102 whereas ErbB2 interacts with Erbin and PICK1. Although, ErbB kinases are concentrated at the NMJ, they are not colocalized with the AChR in cultured muscle cells even in the presence of agrin. Co-expression of PSD-95 causes ErbB4 to form clusters in COS cells. We propose that PDZ domain-containing proteins play a role in anchoring ErbB proteins at the neuromuscular junction, and/or mediating downstream signaling pathways. Such mechanisms could be important for the maintenance and function of the synapse.
Collapse
Affiliation(s)
- Yang Z Huang
- Department of Neurobiology, Pathology, Physical Medicine and Rehabilitation, University of Alabama at Birmingham, 35294-0021, USA
| | | | | | | | | | | |
Collapse
|
39
|
Hu WY, Fukuda N, Kotani M, Kanmatsuse K. Adenovirus-mediated transfer of ribozyme targeting platelet-derived growth factor A-chain mRNA inhibits growth of vascular smooth muscle cells from spontaneously hypertensive rats. J Cardiovasc Pharmacol 2002; 39:858-65. [PMID: 12021580 DOI: 10.1097/00005344-200206000-00011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Platelet-derived growth factor (PDGF) is a potent stimulator of growth of vascular smooth muscle cells (VSMCs). VSMCs from spontaneously hypertensive rats (SHRs) show exaggerated growth and increasingly express PDGF A-chain messenger RNA (mRNA). To examine adenovirus-mediated transfer of a ribozyme targeting the PDGF A-chain mRNA as a possible gene therapy for vascular proliferative diseases, a recombinant adenovirus vector encoding a ribozyme that targets rat PDGF A-chain mRNA (Ad. ribozyme) was designed and synthesized and its effect on the growth of VSMCs from SHRs was investigated. This vector dose-dependently inhibited DNA synthesis in VMSCs from SHRs, whereas an adenovirus vector encoding the Escherichia coli LacZ gene (Ad. LacZ) did not affect DNA synthesis. Ad. ribozyme significantly suppressed proliferation of VSMCs from SHRs in a dose-dependent manner. Ad. LacZ had no effect. Ad. ribozyme significantly inhibited expression of PDGF A-chain mRNA and PDGF-AA protein in VSMCs from SHRs. Ad. LacZ had no effect. These results demonstrated that adenovirus-mediated transfer of a ribozyme targeting the PDGF A-chain mRNA effectively and specifically inhibited the growth of VSMCs from SHRs with suppression of PDGF A-chain mRNA and PDGF-AA protein expression. Adenovirus-mediated transfer of ribozyme targeting PDGF A-chain mRNA may be a feasible gene therapy for vascular proliferative diseases.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cell Division/drug effects
- Cell Division/genetics
- Drug Delivery Systems/methods
- Gene Transfer Techniques/statistics & numerical data
- Genetic Therapy/methods
- Genetic Therapy/statistics & numerical data
- Genetic Vectors/administration & dosage
- Growth Inhibitors/genetics
- Growth Inhibitors/pharmacology
- Growth Inhibitors/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/biosynthesis
- Platelet-Derived Growth Factor/genetics
- RNA, Catalytic/administration & dosage
- RNA, Catalytic/genetics
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Rats, Inbred SHR
Collapse
Affiliation(s)
- Wen-Yang Hu
- Second Department of Internal Medicine, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
40
|
Yotnda P, Chen DH, Chiu W, Piedra PA, Davis A, Templeton NS, Brenner MK. Bilamellar cationic liposomes protect adenovectors from preexisting humoral immune responses. Mol Ther 2002; 5:233-41. [PMID: 11863412 DOI: 10.1006/mthe.2002.0545] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenoviral vectors have been widely used for gene therapy, but they are limited both by the presence of a humoral immune response that dramatically decreases the level of transduction after reinjection and by their requirement for target cells to express appropriate receptors such as Coxsackie adenovirus receptor (CAR). To overcome both limits, we encapsulated adenovectors using bilamellar DOTAP:chol liposomes. Electron micrography (EM) showed that these liposomes efficiently encapsulated the vectors, allowing CAR-independent adenovector transduction of otherwise resistant cells. DOTAP:chol-encapsulated adenovectors encoding LacZ or alpha(1)-antitrypsin inhibitor (AAT) were also functionally resistant ex vivo and in vivo to the neutralizing effects of human anti-adenoviral antibodies, unlike other liposomal systems. Hence, bilamellar DOTAP:chol liposomes may be useful for applications using adenovectors in which the target cells lack adenoviral receptors or in which the recipient already has or develops a neutralizing antibody response that would otherwise inactivate readministered vector.
Collapse
Affiliation(s)
- Patricia Yotnda
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Weld KJ, Mayher BE, Allay JA, Cockroft JL, Reed CP, Randolph MM, Lu Y, Steiner MS, Gingrich JR. Transrectal gene therapy of the prostate in the canine model. Cancer Gene Ther 2002; 9:189-96. [PMID: 11857037 DOI: 10.1038/sj.cgt.7700425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2001] [Indexed: 11/08/2022]
Abstract
Direct transrectal delivery of therapeutic genes utilizing adenoviral vectors for advanced prostate cancer may offer effective treatment at the molecular level. Large animal models to assess feasibility and the intraprostatic and systemic dissemination patterns of these vectors have not been reported. For these studies, a replication-deficient (E1(-)/E3(-)) recombinant adenovirus (AdRSVlacZ) expressing bacterial beta-galactosidase (beta-gal) was delivered under transrectal ultrasound guidance. Two prostate biopsies, followed by concurrent injection of 4.8 x 10(9) pfu of the adenoviral vector divided into either 1 or 2 mL of diluent, were performed (n=4). Swabs of the rectum, sputum, and urine were collected and after 72 hours, the animals were sacrificed. Specimens were assayed for the presence of virus and beta-gal activity. Rectal swabs were transiently positive, whereas urine and sputum samples showed no detectable vector throughout the experiment. Beta-gal activity was observed at the prostate injection sites with detectable activity noted up to 7.5 mm away from the injection site. Systemic dissemination was observed regardless of the injected volume. In conclusion, transrectal prostate biopsy with concurrent prostate injection is a feasible method to deliver therapeutic adenoviral vectors for the treatment of prostate cancer; however, systemic distribution and temporary rectal shedding of virus should be anticipated.
Collapse
Affiliation(s)
- Kyle J Weld
- Urologic Research Laboratories, University of Tennessee, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Schiedner G, Hertel S, Johnston M, Biermann V, Dries V, Kochanek S. Variables affecting in vivo performance of high-capacity adenovirus vectors. J Virol 2002; 76:1600-9. [PMID: 11799154 PMCID: PMC135880 DOI: 10.1128/jvi.76.4.1600-1609.2002] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In high-capacity adenovirus (HC-Ad) vectors the size and/or composition of the vector genome influences vector stability during production and the expression profile following gene transfer. Typically, an HC-Ad vector will contain both a gene or an expression cassette and stuffer DNA that is required to balance the final vector genome to a size of between 27 and 36 kb. To gain an improved understanding of factors that may influence gene expression from HC-Ad vectors, we have generated a series of vectors that carry different combinations of human alpha-1 antitrypsin (hAAT) expression constructs and stuffer DNAs. Expression in vitro did not predict in vivo performance: all vectors expressed hAAT at similar levels when tested in cell culture. Hepatic expression was evaluated following in vivo gene transfer in C57BL/6J mice. hAAT levels obtained from genomic DNA were significantly higher than levels achieved with small cDNA expression cassettes. Expression was independent of the orientation and only marginally influenced by the location of the expression cassette within the vector genome. The use of lambda stuffer DNA resulted in low-level but stable expression for at least 3 months when higher doses were applied. A potential matrix attachment region element was identified within the hAAT gene and caused a 10-fold increase in expression when introduced in an HC-Ad vector genome carrying a phosphoglycerate kinase (pgk) hAAT cDNA construct. We also illustrate the influence of the promoter on anti-hAAT antibody formation in C57BL/6J mice: a human cytomegalovirus but not a pgk promoter resulted in an anti-hAAT antibody response. Thus, the overall design of HC-Ad vectors may significantly influence amounts and duration of gene expression at different levels.
Collapse
Affiliation(s)
- Gudrun Schiedner
- Center for Molecular Medicine (ZMMK), University of Cologne, D-50931 Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Morral N, O'Neal WK, Rice K, Leland MM, Piedra PA, Aguilar-Córdova E, Carey KD, Beaudet AL, Langston C. Lethal toxicity, severe endothelial injury, and a threshold effect with high doses of an adenoviral vector in baboons. Hum Gene Ther 2002; 13:143-54. [PMID: 11779418 DOI: 10.1089/10430340152712692] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The effects of intravenous administration of a first-generation adenoviral vector expressing beta-galactosidase were compared in two baboons receiving a high dose or lower dose of vector, 1.2 x 10(13) or 1.2 x 10(12) particles/kg, respectively. The high-dose baboon developed acute symptoms, decreased platelet counts, and increased liver enzymes, and became moribund at 48 hr after injection, while the lower-dose baboon developed no symptoms. Expression of the beta-galactosidase transgene was prominent in liver, spleen, and endothelium of the arterial vasculature in the high-dose baboon, but was much more limited and spared the endothelium in the lower-dose baboon. Injury to the vascular endothelium was the most prominent abnormality in the high-dose baboon. Extensive histological studies provide a detailed picture of the pathology associated with a lethal dose of first-generation adenoviral vector in a primate.
Collapse
Affiliation(s)
- Núria Morral
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Roberts ML, Athanasopoulos T, Pohlschmidt M, Duisit G, Cosset FL, Dickson G. Post-mitotic, differentiated myotubes efficiently produce retroviral vector from hybrid adeno-retrovirus templates. Gene Ther 2001; 8:1580-6. [PMID: 11704819 DOI: 10.1038/sj.gt.3301555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2001] [Accepted: 07/18/2001] [Indexed: 11/09/2022]
Abstract
We have examined the ability of proliferating myoblasts and post-mitotic, differentiated myotubes to produce retroviral vector using hybrid adeno-retroviral vectors as templates. We show that production of retroviral vector from myoblasts peaks 48 h after adenoviral infection at 4.8 x 10(4) cfu/ml and is scarcely detectable by 96 h. Both fully and partially differentiated myotubes were able to generate a sustained increase in the levels of retroviral vector compared with myoblasts peaking 48 h at 1.4 x 10(5) cfu/ml and 1.8 x 10(5) cfu/ml, respectively. Addition of the cell cycle inhibitor aphidicolin (5 microg/ml) had no effect on the production of retroviral vector from fully differentiated myotubes, but resulted in an 80% increase in vector production from partially differentiated myotubes. Thus indicating that retroviral vector production is more efficient in post-mitotic myotubes and is independent of muscle cell cycle progression.
Collapse
Affiliation(s)
- M L Roberts
- Division of Biochemistry, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey, UK
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Donor myoblast migration is a major limiting factor in the success of myoblast transfer therapy, a potential treatment for Duchenne muscular dystrophy. A possible strategy to promote the migration of donor myoblasts into host muscle is to enhance their proliferation and delay their fusion, two properties that are major characteristics of myoblasts in regenerating skeletal muscle in MyoD null (-/-) mice. Here we investigate whether the migration of MyoD (-/-) donor myoblasts into host muscle is enhanced in vivo. Sliced muscle grafts from male MyoD (-/-) or normal control (Balb/c) mice were transplanted into the muscles of female normal (Balb/c) host mice. Muscles were sampled at 1, 3, and 12 weeks after grafting, and the fate of male donor myoblasts within female host muscles determined by in situ hybridization with the mouse Y-chromosome-specific Y-1 probe. MyoD (-/-) donor myoblasts migrated into host muscle continuously over 1, 3, and 12 weeks after grafting, in contrast with Balb/c donor myoblasts, whose overall numbers and migratory distances did not increase significantly after 1 week. These results strongly support a role for elevated donor myoblast proliferation and/or their delayed fusion in enhancing migration into host muscle in vivo, and endorse the use of either genetically engineered donor myoblasts, or the administration of exogenous myoblast mitogens to improve donor myoblast migration in myoblast transfer therapy.
Collapse
Affiliation(s)
- G M Smythe
- Department of Anatomy and Human Biology, The University of Western Australia, Crawley, Perth, 6009, Western Australia
| | | |
Collapse
|
46
|
Zoltick PW, Chirmule N, Schnell MA, Gao GP, Hughes JV, Wilson JM. Biology of E1-deleted adenovirus vectors in nonhuman primate muscle. J Virol 2001; 75:5222-9. [PMID: 11333904 PMCID: PMC114928 DOI: 10.1128/jvi.75.11.5222-5229.2001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2000] [Accepted: 02/19/2001] [Indexed: 11/20/2022] Open
Abstract
Adenovirus vectors have been studied as vehicles for gene transfer to skeletal muscle, an attractive target for gene therapies for inherited and acquired diseases. In this setting, immune responses to viral proteins and/or transgene products cause inflammation and lead to loss of transgene expression. A few studies in murine models have suggested that the destructive cell-mediated immune response to virally encoded proteins of E1-deleted adenovirus may not contribute to the elimination of transgene-expressing cells. However, the impact of immune responses following intramuscular administration of adenovirus vectors on transgene stability has not been elucidated in larger animal models such as nonhuman primates. Here we demonstrate that intramuscular administration of E1-deleted adenovirus vector expressing rhesus monkey erythropoietin or growth hormone to rhesus monkeys results in generation of a Th1-dependent cytotoxic T-cell response to adenovirus proteins. Transgene expression dropped significantly over time but was still detectable in some animals after 6 months. Systemic levels of adenovirus-specific neutralizing antibodies were generated, which blocked vector readministration. These studies indicate that the cellular and humoral immune response generated to adenovirus proteins, in the context of transgenes encoding self-proteins, hinders long-term transgene expression and readministration with first-generation vectors.
Collapse
Affiliation(s)
- P W Zoltick
- Institute for Human Gene Therapy, University of Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
47
|
Zhou H, Zhao T, Pastore L, Nageh M, Zheng W, Rao XM, Beaudet AL. A Cre-expressing cell line and an E1/E2a double-deleted virus for preparation of helper-dependent adenovirus vector. Mol Ther 2001; 3:613-22. [PMID: 11319924 DOI: 10.1006/mthe.2001.0288] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenoviral vectors are attractive for the delivery of transgenes into mammalian cells because of their efficient transduction, high titer, and stability. The major concerns with using E1-deleted adenoviral vectors in gene therapy are the pathogenic potential of the virus backbone and the leaky viral protein synthesis that leads to host immune responses and a short duration of transgene expression. Helper-dependent (HD) adenoviral vectors that are devoid of all viral protein-coding sequences have significantly increased the safety and reduced the immunogenicity of these vectors. Currently available HD vectors depend on an E1-deleted adenovirus as a helper to provide viral proteins in trans. As a consequence, contamination with helper virus cannot be avoided in the HD vector preparation though it can be decreased to 0.01% using a Cre/loxP mechanism. Since the presence of E1-deleted helper virus may have substantial unwanted effects, we have developed a new Cre-expressing cell line based on an E1- and E2a-complementing cell. This new cell line can efficiently cleave the packaging region in the helper virus genome. We have also developed an E1 and E2a double-deleted helper virus. By using the CreE cell with the helper virus deleted in both the E1 and the E2a genes it may be possible to further improve the safety of the vectors.
Collapse
Affiliation(s)
- H Zhou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- J M Wilson
- Institute for Human Gene Therapy, The University of Pennsylvania and the Wistar Institute, Philadelphia, PA 19104-4268, USA
| |
Collapse
|
49
|
Kimura E, Maeda Y, Arima T, Nishida Y, Yamashita S, Hara A, Uyama E, Mita S, Uchino M. Efficient repetitive gene delivery to skeletal muscle using recombinant adenovirus vector containing the Coxsackievirus and adenovirus receptor cDNA. Gene Ther 2001; 8:20-7. [PMID: 11402298 DOI: 10.1038/sj.gt.3301359] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2000] [Accepted: 10/11/2000] [Indexed: 11/09/2022]
Abstract
To improve adenovirus-mediated gene delivery to skeletal muscle, we have used a recombinant adenovirus vector encoding the human Coxsackievirus and adenovirus receptor (hCAR). Because CAR is expressed at a lower level in rodent myoblasts and muscle fibers than in other tissues, we expected that elevated expression of CAR in skeletal muscle would improve the efficacy of adenovirus-mediated gene transfer. Since the mouse myoblasts, C2C12 cells, showed low sensitivity to infection by recombinant adenovirus 5, we initially infected these cells at a high multiplicity of infection (MOI) of 250 with the recombinant adenovirus containing hCAR cDNA and LacZ gene. Subsequent infection by recombinant adenovirus containing the marker gene, green fluorescence protein, became efficient even at a low MOI of 25. Thus, elevated hCAR expression in mouse muscle fibers made a second virus inoculation at low doses possible. We also demonstrated that the elevated hCAR expression did not influence muscle membrane integrity. Our results suggest that co-expression of CAR and a therapeutic gene by adenovirus vector constitutes a novel strategy to advance gene therapy for hereditary muscle diseases.
Collapse
Affiliation(s)
- E Kimura
- Department of Neurology, Kumamoto University School of Medicine, 1-1-1 Honjo, Kumamoto 860-0811 Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tanabe KK, Cusack JC. Gene Therapy. Surgery 2001. [DOI: 10.1007/978-3-642-57282-1_86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|