1
|
Xu YP, Zhou HY, Wang GC, Zhang Y, Yang T, Zhao Y, Li RT, Zhang RR, Guo Y, Wang X, Li XF, Qin CF, Tang R. Rational Design of a Replication-Competent and Inheritable Magnetic Viruses for Targeting Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002435. [PMID: 32954651 DOI: 10.1002/smll.202002435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/20/2020] [Indexed: 06/11/2023]
Abstract
Infection with live-attenuated vaccines always inevitably induces side effects that reduce their safety. This study suggests a concept of magnetic virus produced by genetically modifying viral surfaces with Fe3 O4 nanoparticles (NPs) to control their tropisms. An iron-affinity peptide is designed to be displayed on the viral surface protein (VP1) of human enterovirus type 71 (EV71), a typical nonenveloped picornavirus, as the model. The modified EV71 can self-bind with Fe3 O4 NPs under physiological conditions, resulting in novel EV71-Fe3 O4 hybrid materials. This rationally engineered EV71 with Fe3 O4 retains its original biological infectivity, but its tropism can be precisely controlled by magnetism. Both in vitro and in vivo experiments demonstrate that EV71-Fe3 O4 can infect only a desired area within the limit of the applied magnetic field, which effectively reduces its pathological damage. More importantly, this characteristic of EV71 can be inherited due to the gene-induced coassembly of viruses and NPs. This achievement provides a proof of concept in virus vaccine improvement by a combination of gene modification and material incorporation, leading to great potential for biomedical developments.
Collapse
Affiliation(s)
- Yan-Peng Xu
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hang-Yu Zhou
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
- Suzhou Institute of System Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215000, China
| | - Guang-Chuan Wang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Ying Zhang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Tianxu Yang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Yueqi Zhao
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Rui-Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Xiaoyu Wang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Ruikang Tang
- Center for Biomaterials and Biopathways Depart of Chemistry, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang Province, 310027, China
| |
Collapse
|
2
|
Carriere J, Rao Y, Liu Q, Lin X, Zhao J, Feng P. Post-translational Control of Innate Immune Signaling Pathways by Herpesviruses. Front Microbiol 2019; 10:2647. [PMID: 31798565 PMCID: PMC6868034 DOI: 10.3389/fmicb.2019.02647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
Herpesviruses constitute a large family of disease-causing DNA viruses. Each herpesvirus strain is capable of infecting particular organisms with a specific cell tropism. Upon infection, pattern recognition receptors (PRRs) recognize conserved viral features to trigger signaling cascades that culminate in the production of interferons and pro-inflammatory cytokines. To invoke a proper immune response while avoiding collateral tissue damage, signaling proteins involved in these cascades are tightly regulated by post-translational modifications (PTMs). Herpesviruses have developed strategies to subvert innate immune signaling pathways in order to ensure efficient viral replication and achieve persistent infection. The ability of these viruses to control the proteins involved in these signaling cascades post-translationally, either directly via virus-encoded enzymes or indirectly through the deregulation of cellular enzymes, has been widely reported. This ability provides herpesviruses with a powerful tool to shut off or restrict host antiviral and inflammatory responses. In this review, we highlight recent findings on the herpesvirus-mediated post-translational control along PRR-mediated signaling pathways.
Collapse
Affiliation(s)
| | | | | | | | | | - Pinghui Feng
- Section of Infection and Immunity, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
3
|
Ostrowski RA, Metgud S, Tehrani R, Jay WM. Varicella Zoster Virus in Giant Cell Arteritis: A Review of Current Medical Literature. Neuroophthalmology 2019; 43:159-170. [PMID: 31312240 DOI: 10.1080/01658107.2019.1604763] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/30/2019] [Accepted: 04/04/2019] [Indexed: 10/26/2022] Open
Abstract
In recent years, the search for the cause of giant cell arteritis (GCA) has led investigators to look to varicella zoster virus (VZV) as the answer. In some ways, the nature of VZV infection makes it an attractive explanation for the pathology observed in GCA. However, studies to date yield a level of inconsistency that still leaves uncertainty as to whether VZV directly causes GCA, and positive findings have not been successfully reproduced.
Collapse
Affiliation(s)
- Rochella A Ostrowski
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Loyola, University Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| | - Sheela Metgud
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Loyola, University Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| | - Rodney Tehrani
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Loyola, University Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| | - Walter M Jay
- Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois, USA
| |
Collapse
|
4
|
Contributions of the glycocalyx, endothelium, and extravascular compartment to the blood-brain barrier. Proc Natl Acad Sci U S A 2018; 115:E9429-E9438. [PMID: 30217895 DOI: 10.1073/pnas.1802155115] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The endothelial cells that form the blood-brain barrier (BBB) are coated with glycocalyx, on the luminal side, and with the basement membrane and astrocyte endfeet, on the abluminal side. However, it is unclear how exactly the glycocalyx and extravascular structures contribute to BBB properties. We used two-photon microscopy in anesthetized mice to record passive transport of four different-sized molecules-sodium fluorescein (376 Da), Alexa Fluor (643 Da), 40-kDa dextran, and 150-kDa dextran-from blood to brain, at the level of single cortical capillaries. Both fluorescein and Alexa penetrated nearly the entire glycocalyx volume, but the dextrans penetrated less than 60% of the volume. This suggested that the glycocalyx was a barrier for large but not small molecules. The estimated permeability of the endothelium was the same for fluorescein and Alexa but several-fold lower for the larger dextrans. In the extravascular compartment, co-localized with astrocyte endfeet, diffusion coefficients of the dyes were an order of magnitude lower than in the brain parenchyma. This suggested that the astrocyte endfeet and basement membrane also contributed to BBB properties. In conclusion, the passive transport of small and large hydrophilic molecules through the BBB was determined by three separate barriers: the glycocalyx, the endothelium, and the extravascular compartment. All three barriers must be taken into account in drug delivery studies and when considering BBB dysfunction in disease states.
Collapse
|
5
|
Buckingham EM, Foley MA, Grose C, Syed NA, Smith ME, Margolis TP, Thurtell MJ, Kardon R. Identification of Herpes Zoster-Associated Temporal Arteritis Among Cases of Giant Cell Arteritis. Am J Ophthalmol 2018; 187:51-60. [PMID: 29294312 PMCID: PMC5866091 DOI: 10.1016/j.ajo.2017.12.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE To examine whether herpes zoster antigen (also called varicellazoster virus antigen) was detectable in temporal artery biopsies taken from individuals with giant cell arteritis (GCA). DESIGN Retrospective comparative case series. METHODS Sections of formalin-fixed paraffin-embedded temporal arteries were examined first by hematoxylin-eosin (H&E) staining to establish the diagnosis of GCA. Adjacent sections of the same biopsy were then examined by immunohistochemistry, using 2 different monoclonal antibodies against a major antigen of varicella-zoster virus called gE. Pathologic specimens were obtained from patients cared for at the University of Iowa and Washington University in St. Louis ophthalmology clinics. RESULTS The study included biopsies from 25 patients with symptoms of GCA as well as positive H&E pathology and 25 patients with symptoms compatible with GCA but negative H&E pathology. Among the GCA-positive group, 3 patients had positive staining for herpes zoster antigen. Among the GCA-negative group, herpes zoster antigen was not detected in any biopsy. In both groups of patients, false-positive staining for herpes zoster antigen was detected in the presence of calcifications in the arteries. False-positive staining was also detected on some extra-arterial skeletal muscle and erythrocytes. CONCLUSION Herpes zoster antigen was detected in 3 of 25 temporal arteries from patients with biopsy-proven GCA. One of the 3 positive cases was noteworthy because the patient had had herpes zoster ophthalmicus diagnosed 3 weeks before the onset of GCA symptoms. False-positive staining for herpes zoster antigen was detected on several temporal artery biopsies.
Collapse
|
6
|
Kasemkijwattana C, Menetrey J, Somogyl G, Moreland MS, Fu FH, Buranapanitkit B, Watkins SC, Huard J. Development of Approaches to Improve the Healing following Muscle Contusion. Cell Transplant 2017; 7:585-98. [PMID: 9853587 DOI: 10.1177/096368979800700609] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Muscle injuries are a challenging problem in traumatology, and the most frequent occurrence in sports medicine. Muscle contusions are among the most common muscle injuries. Although this injury is capable of healing, an incomplete functional recovery often occurs, depending on the severity of the blunt trauma. We have developed an animal model of muscle contusion in mice (high energy blunt trauma) and characterized the muscle's ability to heal following this injury using histology and immunohistochemistry to determine the level of muscle regeneration and the development of scar tissue. We have observed a massive muscle regeneration occurring in the first 2 wk postinjury that is subsequently followed by the development of muscle fibrosis. Based on these observations, we propose that the enhancement of muscle growth and regeneration, as well as the prevention of fibrotic development, could be used as approach(es) to improve the healing of muscle injuries. In fact, we have identified three growth factors (bFGF, IGF-1, and NGF) capable of enhancing myoblast proliferation and differentiation in vitro and improving the healing of the injured muscle in vivo. Furthermore, the ability of adenovirus to mediate direct and ex vivo gene transfer of β-galactosidase into the injured site opens possibilities of delivering an efficient and persistent expression of these growth factors in the injured muscle. These studies should help in the development of strategies to promote efficient muscle healing with complete functional recovery following muscle contusion. © 1998 Elsevier Science Inc.
Collapse
Affiliation(s)
- C Kasemkijwattana
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh, and Children's Hospital of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Primary Murine Myotubes as a Model for Investigating Muscular Dystrophy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:594751. [PMID: 26380282 PMCID: PMC4561302 DOI: 10.1155/2015/594751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 03/11/2015] [Indexed: 11/18/2022]
Abstract
Muscular dystrophies caused by defects in various genes are often associated with impairment of calcium homeostasis. Studies of calcium currents are hampered because of the lack of a robust cellular model. Primary murine myotubes, formed upon satellite cell fusion, were examined for their utilization as a model of adult skeletal muscle. We enzymatically isolated satellite cells and induced them to differentiation to myotubes. Myotubes displayed morphological and physiological properties resembling adult muscle fibers. Desmin and myosin heavy chain immunoreactivity in the differentiated myotubes were similar to the mature muscle cross-striated pattern. The myotubes responded to electrical and chemical stimulations with sarcoplasmic reticulum calcium release. Presence of L-type calcium channels in the myotubes sarcolemma was confirmed via whole-cell patch-clamp technique. To assess the use of myotubes for studying functional mutation effects lentiviral transduction was applied. Satellite cells easily underwent transduction and were able to retain a positive expression of lentivirally encoded GFP up to and after the formation of myotubes, without changes in their physiological and morphological properties. Thus, we conclude that murine myotubes may serve as a fruitful cell model for investigating calcium homeostasis in muscular dystrophy and the effects of gene modifications can be assessed due to lentiviral transduction.
Collapse
|
8
|
Infection of myofibers contributes to increased pathogenicity during infection with an epidemic strain of chikungunya virus. J Virol 2013; 88:2414-25. [PMID: 24335291 DOI: 10.1128/jvi.02716-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitoes that is known to cause severe arthritis and myositis in affected patients. The ongoing epidemic began in eastern Africa in 2004 and then spread to islands of the Indian Ocean, India, and Southeast Asia, ultimately afflicting millions. During this outbreak, more severe disease manifestations, including fatalities, have been documented. The reasons for this change in pathogenesis are multifactorial but likely include mutations that have arisen in the viral genome which could alter disease pathogenesis. To test this hypothesis, we used a murine model of CHIKV to compare the disease pathogeneses of two recombinant strains of CHIKV, the first derived from the La Reunion outbreak in 2006 (LR2006 OPY1) and the second isolated from Senegal in 1983 (37997). While the two strains exhibited similar growth in mammalian cells in vitro, we observed more severe clinical disease and pathology in mice infected with the LR2006 OPY1 strain of CHIKV, which included prolonged viremia and elevated viral titers and persistence in the muscle, resulting in devastating myonecrosis. Both CHIKV strains infected connective tissue fibroblasts of the muscle, but only the LR2006 OPY1 strain replicated within myofibers in vivo, despite similar growth of the two strains in these cell types in vitro. However, when the 37997 strain was administered directly into muscle, myofiber infection was comparable to that in LR2006 OPY1-infected mice. These results indicate that differences in the ability of the strain of CHIKV to establish infection in myofibers may contribute to the increased disease severity. IMPORTANCE CHIKV is an emerging pathogen that causes significant morbidity. Little is known about the pathogenesis of the disease, and this study suggests that the ability of a recent epidemic strain to infect myofibers results in increased disease severity. Better understanding of how CHIKV causes disease contributes to the ultimate goal of creating therapeutics to alleviate the impact of this debilitating virus.
Collapse
|
9
|
Torricelli AAM, Singh V, Santhiago MR, Wilson SE. The corneal epithelial basement membrane: structure, function, and disease. Invest Ophthalmol Vis Sci 2013; 54:6390-400. [PMID: 24078382 DOI: 10.1167/iovs.13-12547] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The corneal epithelial basement membrane (BM) is positioned between basal epithelial cells and the stroma. This highly specialized extracellular matrix functions not only to anchor epithelial cells to the stroma and provide scaffolding during embryonic development but also during migration, differentiation, and maintenance of the differentiated epithelial phenotype. Basement membranes are composed of a diverse assemblage of extracellular molecules, some of which are likely specific to the tissue where they function; but in general they are composed of four primary components--collagens, laminins, heparan sulfate proteoglycans, and nidogens--in addition to other components such as thrombospondin-1, matrilin-2, and matrilin-4 and even fibronectin in some BM. Many studies have focused on characterizing BM due to their potential roles in normal tissue function and disease, and these structures have been well characterized in many tissues. Comparatively few studies, however, have focused on the function of the epithelial BM in corneal physiology. Since the normal corneal stroma is avascular and has relatively low keratocyte density, it is expected that the corneal BM would be different from the BM in other tissues. One function that appears critical in homeostasis and wound healing is the barrier function to penetration of cytokines from the epithelium to stroma (such as transforming growth factor β-1), and possibly from stroma to epithelium (such as keratinocyte growth factor). The corneal epithelial BM is also involved in many inherited and acquired corneal diseases. This review examines this structure in detail and discusses the importance of corneal epithelial BM in homeostasis, wound healing, and disease.
Collapse
|
10
|
Pasut A, Jones AE, Rudnicki MA. Isolation and culture of individual myofibers and their satellite cells from adult skeletal muscle. J Vis Exp 2013:e50074. [PMID: 23542587 PMCID: PMC3639710 DOI: 10.3791/50074] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Muscle regeneration in the adult is performed by resident stem cells called satellite cells. Satellite cells are defined by their position between the basal lamina and the sarcolemma of each myofiber. Current knowledge of their behavior heavily relies on the use of the single myofiber isolation protocol. In 1985, Bischoff described a protocol to isolate single live fibers from the Flexor Digitorum Brevis (FDB) of adult rats with the goal to create an in vitro system in which the physical association between the myofiber and its stem cells is preserved 1. In 1995, Rosenblattmodified the Bischoff protocol such that myofibers are singly picked and handled separately after collagenase digestion instead of being isolated by gravity sedimentation 2, 3. The Rosenblatt or Bischoff protocol has since been adapted to different muscles, age or conditions 3-6. The single myofiber isolation technique is an indispensable tool due its unique advantages. First, in the single myofiber protocol, satellite cells are maintained beneath the basal lamina. This is a unique feature of the protocol as other techniques such as Fluorescence Activated Cell Sorting require chemical and mechanical tissue dissociation 7. Although the myofiber culture system cannot substitute for in vivo studies, it does offer an excellent platform to address relevant biological properties of muscle stem cells. Single myofibers can be cultured in standard plating conditions or in floating conditions. Satellite cells on floating myofibers are subjected to virtually no other influence than the myofiber environment. Substrate stiffness and coating have been shown to influence satellite cells' ability to regenerate muscles 8, 9 so being able to control each of these factors independently allows discrimination between niche-dependent and -independent responses. Different concentrations of serum have also been shown to have an effect on the transition from quiescence to activation. To preserve the quiescence state of its associated satellite cells, fibers should be kept in low serum medium 1-3. This is particularly useful when studying genes involved in the quiescence state. In serum rich medium, satellite cells quickly activate, proliferate, migrate and differentiate, thus mimicking the in vivo regenerative process 1-3. The system can be used to perform a variety of assays such as the testing of chemical inhibitors; ectopic expression of genes by virus delivery; oligonucleotide based gene knock-down or live imaging. This video article describes the protocol currently used in our laboratory to isolate single myofibers from the Extensor Digitorum Longus (EDL) muscle of adult mice (6-8 weeks old).
Collapse
Affiliation(s)
- Alessandra Pasut
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute
| | | | | |
Collapse
|
11
|
HSV-1 amplicon vectors launch the production of heterologous rotavirus-like particles and induce rotavirus-specific immune responses in mice. Mol Ther 2012; 20:1810-20. [PMID: 22713696 DOI: 10.1038/mt.2012.108] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Virus-like particles (VLPs) are promising vaccine candidates because they represent viral antigens in the authentic conformation of the virion and are therefore readily recognized by the immune system. As VLPs do not contain genetic material they are safer than attenuated virus vaccines. In this study, herpes simplex virus type 1 (HSV-1) amplicon vectors were constructed to coexpress the rotavirus (RV) structural genes VP2, VP6, and VP7 and were used as platforms to launch the production of RV-like particles (RVLPs) in vector-infected mammalian cells. Despite the observed splicing of VP6 RNA, full-length VP6 protein and RVLPs were efficiently produced. Intramuscular injection of mice with the amplicon vectors as a two-dose regimen without adjuvants resulted in RV-specific humoral immune responses and, most importantly, immunized mice were partially protected at the mucosal level from challenge with live wild-type (wt) RV. This work provides proof of principle for the application of HSV-1 amplicon vectors that mediate the efficient production of heterologous VLPs as genetic vaccines.
Collapse
|
12
|
Pisconti A, Bernet JD, Olwin BB. Syndecans in skeletal muscle development, regeneration and homeostasis. Muscles Ligaments Tendons J 2012; 2:1-9. [PMID: 23738267 PMCID: PMC3666495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Skeletal muscle is a highly dynamic tissue that can change in size in response to physiological demands and undergo successful regeneration even upon extensive injury. A population of resident stem cells, termed satellite cells, accounts for skeletal muscle plasticity, maintenance and regeneration. Mammalian satellite cells, generated from muscle precursor cells during development, are maintained quiescent in the musculature throughout a lifespan, but ready to activate, proliferate and differentiate into myocytes upon demand. Syndecans are transmembrane heparan sulfate proteoglycans expressed in muscle precursors during embryonic development and in satellite cells during postnatal life. In the last decades a number of crucial functions for syndecans in myogenesis and muscle disease have been described. Here we review the current knowledge of the multiple roles played by syndecans in the skeletal muscle of several animal models and explore future perspectives for human muscle health, with a focus on muscle aging and muscular dystrophy.
Collapse
Affiliation(s)
- Addolorata Pisconti
- Corresponding author: Addolorata Pisconti, MCD-Biology, 347 UCB, Boulder, CO 80309, USA, e-mail:
| | | | | |
Collapse
|
13
|
Lee CW, Fukushima K, Usas A, Xin L, Pelinkovic D, Martinek V, Somogyi G, Robbins PD, Fu FH, Huard J. BIOLOGICAL INTERVENTION BASED ON CELL AND GENE THERAPY TO IMPROVE MUSCLE HEALING AFTER LACERATION. ACTA ACUST UNITED AC 2011. [DOI: 10.1142/s0218957700000264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Muscle laceration is a challenging problem in traumatology and is common in sports injuries, with functional recovery remaining slow and incomplete. Even though muscles retain their ability to regenerate after injury, muscles' healing process after such injuries has been found to be very slow and often leads to incomplete muscle recovery. Growth factors may have a role in enhancing recovery. Our previous study showed that IGF-1, β-FGF and NGF can improve myoblast proliferation and differentiation in vitro. We then investigated whether the delivery of IGF-1 would improve muscle healing after injuries. We observed that muscle regeneration was enhanced in lacerated muscles treated with IGF-1 protein, which consequently led to an improvement in muscle healing. However, the rapid clearance and short biological half-lives of these proteins may have limited the success of this approach. We then investigated the efficiency of gene therapy based on adenovirus to deliver a stable expression of the growth factor IGF-1. Although a slight improvement in the healing process occurred in the muscle injected with adenovirus (AIGF), the combination of myoblast transplantation and gene therapy with the ex vivo approach further improved the healing process. The injection of normal myoblasts into the injured muscle led to the best improvement of muscle healing at two weeks post-injection. Implantation of normal minced muscle into mdx mice was also capable of improving muscle healing at 2–4 weeks post-implantation. These studies will further our understanding of muscle healing post-injury and help in the development of strategies to promote efficient muscle healing and complete functional recovery after common muscle injuries.
Collapse
Affiliation(s)
- Chang Woo Lee
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Kazumasa Fukushima
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Arvydas Usas
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Lin Xin
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Dalip Pelinkovic
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Vladimir Martinek
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - George Somogyi
- Department of Pharmacology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Paul D. Robbins
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Freddie H. Fu
- Department of Orthopaedic Surgery, Division of Sports Medicine, Athletic Department, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, University of Pittsburgh and Children's Hospital of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
14
|
Koppanati BM, Li J, Reay DP, Wang B, Daood M, Zheng H, Xiao X, Watchko JF, Clemens PR. Improvement of the mdx mouse dystrophic phenotype by systemic in utero AAV8 delivery of a minidystrophin gene. Gene Ther 2010; 17:1355-62. [PMID: 20535217 PMCID: PMC2939256 DOI: 10.1038/gt.2010.84] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating primary muscle disease with pathological changes in skeletal muscle that are ongoing at the time of birth. Progressive deterioration in striated muscle function in affected individuals ultimately results in early death due to cardio-pulmonary failure. As affected individuals can be identified before birth by prenatal genetic testing for DMD, gene replacement treatment can be started in utero. This approach offers the possibility of preventing pathological changes in muscle that begin early in life. To test in utero gene transfer in the mdx mouse model of DMD, a minidystrophin gene driven by the human cytomegalovirus promoter was delivered systemically by an intraperitoneal injection to the fetus at embryonic day 16. Treated mdx mice studied at 9 weeks after birth showed widespread expression of recombinant dystrophin in skeletal muscle, restoration of the dystrophin-associated glycoprotein complex in dystrophin-expressing muscle fibers, improved muscle pathology, and functional benefit to the transduced diaphragm compared with untreated littermate controls. These results support the potential of the AAV8 vector to efficiently cross the blood vessel barrier to achieve systemic gene transfer to skeletal muscle in utero in a mouse model of muscular dystrophy, to significantly improve the dystrophic phenotype and to ameliorate the processes that lead to exhaustion of the skeletal muscle regenerative capacity.
Collapse
Affiliation(s)
- B M Koppanati
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Harrison RL, Bonning BC. Proteases as insecticidal agents. Toxins (Basel) 2010; 2:935-53. [PMID: 22069618 PMCID: PMC3153225 DOI: 10.3390/toxins2050935] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 04/26/2010] [Accepted: 04/30/2010] [Indexed: 11/16/2022] Open
Abstract
Proteases from a variety of sources (viruses, bacteria, fungi, plants, and insects) have toxicity towards insects. Some of these insecticidal proteases evolved as venom components, herbivore resistance factors, or microbial pathogenicity factors, while other proteases play roles in insect development or digestion, but exert an insecticidal effect when over-expressed from genetically engineered plants or microbial pathogens. Many of these proteases are cysteine proteases, although insect-toxic metalloproteases and serine proteases have also been examined. The sites of protease toxic activity range from the insect midgut to the hemocoel (body cavity) to the cuticle. This review discusses these insecticidal proteases along with their evaluation and use as potential pesticides.
Collapse
Affiliation(s)
- Robert L. Harrison
- Invasive Insect Biocontrol and Behavior Laboratory, USDA Agricultural Research Service, Plant Sciences Institute, 10300 Baltimore Avenue, Beltsville, Maryland 20705, USA; Robert.L.
| | - Bryony C. Bonning
- Department of Entomology, Iowa State University, 418 Science II, Ames, IA 50011-3222, USA
- Author to whom correspondence should be addressed; ; Tel.: +01-515-294-1989; Fax: +01-515-294-5957
| |
Collapse
|
16
|
Role of the corneal epithelial basement membrane in ocular defense against Pseudomonas aeruginosa. Infect Immun 2009; 77:3264-71. [PMID: 19506010 DOI: 10.1128/iai.00111-09] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa can invade corneal epithelial cells and translocates multilayered corneal epithelia in vitro, but it does not penetrate the intact corneal epithelium in vivo. In healthy corneas, the epithelium is separated from the underlying stroma by a basement membrane containing extracellular matrix proteins and pores smaller than bacteria. Here we used in vivo and in vitro models to investigate the potential of the basement membrane to defend against P. aeruginosa. Transmission electron microscopy of infected mouse corneas in vivo showed penetration of the stroma by P. aeruginosa only where the basement membrane was visibly disrupted by scratch injury, suggesting that the intact basement membrane prevented penetration. This hypothesis was explored using an in vitro Matrigel Transwell model to mimic the corneal basement membrane. P. aeruginosa translocation of multilayered corneal epithelia grown on Matrigel was approximately 100-fold lower than that of cells grown without Matrigel (P < 0.005, t test). Matrigel did not increase transepithelial resistance. Matrigel-grown cells blocked translocation by a P. aeruginosa protease mutant. Without cells, Matrigel also reduced traversal of P. aeruginosa and the protease mutant. Fluorescence microscopy revealed a relative accumulation of bacteria at the superficial epithelium of cells grown on Matrigel at 3 h compared to cells grown on uncoated filters. By 5 h, bacteria accumulated beneath the cells, suggesting direct trapping by the Matrigel. These findings suggest that the basement membrane helps defend the cornea against infection via physical barrier effects and influences on the epithelium and that these roles could be compromised by P. aeruginosa proteases.
Collapse
|
17
|
Reay DP, Bilbao R, Koppanati BM, Cai L, O'Day TL, Jiang Z, Zheng H, Watchko JF, Clemens PR. Full-length dystrophin gene transfer to the mdx mouse in utero. Gene Ther 2008; 15:531-6. [PMID: 18273052 DOI: 10.1038/gt.2008.8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In utero gene therapy for genetic diseases, such as muscular dystrophies, offers potential advantages over postnatal treatment including vector delivery at the earliest point in the disease and treatment prior to full maturation of the immune system. This study examines in utero gene delivery of full-length murine dystrophin to the murine mdx model for Duchenne muscular dystrophy using a high-capacity adenoviral vector. We examined dystrophin expression, spread of vector, morphology and specific force production of the tibialis anterior muscle 9 weeks after intramuscular in utero injection. Recombinant dystrophin was expressed in the hindlimb muscles, with the majority of animals having expression in two muscles of the injected hindlimb. The dystrophin-glycoprotein complex was restored in those muscle fibers expressing recombinant dystrophin. Analysis of the percentage of dystrophin-expressing muscle fibers with centrally placed nuclei revealed effective protection from cycles of degeneration and regeneration normally seen in muscle fibers lacking dystrophin. However, due to low levels of muscle gene transfer, further advances in the efficiency of adenoviral vector-mediated gene delivery would be required for clinical applications of in utero gene therapy for primary myopathies such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- D P Reay
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Shen W, Li Y, Huard J. Musculoskeletal gene therapy and its potential use in the treatment of complicated musculoskeletal infection. Infect Dis Clin North Am 2006; 19:1007-22. [PMID: 16297745 DOI: 10.1016/j.idc.2005.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tissue repair is a major issue in orthopedics. Many musculoskeletal tissues, including cartilage, meniscus, and the anterior cruciate ligament, heal poorly after injury. Recent studies have led to the identification of numerous growth factors and other gene products that can promote the regeneration of damaged musculoskeletal tissues. In the last century, the discovery and evolving use of antibiotics has significantly decreased the prevalence and severity of infectious diseases. In many orthopedic scenarios, however, treatment of infections can be difficult, and often involves a prolonged course of antibiotics with concomitant surgical interventions and loss of tissue. Although studies have demonstrated the successful transfer of target genes and the associated manipulation of the musculoskeletal tissue environment, researchers have made few attempts designed to use gene therapy to treat infectious musculoskeletal diseases in animal models. Before it is possible to use gene-based approaches to treat such diseases effectively, researchers must perform more studies to investigate the potential problems that may arise when using gene therapy in an infectious environment.
Collapse
Affiliation(s)
- Wei Shen
- Growth and Development Laboratory of Children's Hospital of Pittsburgh, 4100 Rangos Research Center, Pittsburgh, PA 15213-2583, USA
| | | | | |
Collapse
|
20
|
Lee H, Koehler DR, Pang CY, Levine RH, Ng P, Palmer DJ, Quinton PM, Hu J. Gene delivery to human sweat glands: a model for cystic fibrosis gene therapy. Gene Ther 2006; 12:1752-60. [PMID: 16034452 DOI: 10.1038/sj.gt.3302587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene therapy vectors are mostly studied in cultured cells, rodents, and sometimes in non-human primates, but it is useful to test them in human tissue prior to clinical trials. In this study, we investigated the possibility of using human sweat glands as a model for testing cystic fibrosis (CF) gene therapy vectors. Human sweat glands are relatively easy to obtain from skin biopsy, and can be tested for CFTR function. Using patients' sweat glands could provide a safe model to study the efficacy of CF gene therapy. As the first step to explore using sweat glands as a model for CF gene therapy, we examined various ex vivo gene delivery methods for a helper-dependent adenovirus (HD-Ad) vector. Gene delivery to sweat glands in skin organ culture was studied by topical application, intradermal injection or submerged culture. We found that transduction efficiency can be enhanced by pretreating isolated sweat glands with dispase, which suggests that the basement membrane is a critical barrier to gene delivery by adenoviral vectors. Using this approach, we showed that Cftr could be efficiently delivered to and expressed by the epithelial cells of sweat glands with our helper-dependent adenoviral vector containing cytokeratin 18 regulatory elements. Based on this study we propose that sweat glands might be used as an alternative model to study CF gene therapy in humans.
Collapse
Affiliation(s)
- H Lee
- Research Institute, The Hospital for Sick Children, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Breshears MA, Eberle R, Ritchey JW. Temporal progression of viral replication and gross and histological lesions in Balb/c mice inoculated epidermally with Saimiriine herpesvirus 1 (SaHV-1). J Comp Pathol 2005; 133:103-13. [PMID: 15964589 DOI: 10.1016/j.jcpa.2005.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Accepted: 01/25/2005] [Indexed: 11/27/2022]
Abstract
Saimiriine herpesvirus 1 (SaHV-1), an alphaherpesvirus enzootic in squirrel monkeys, is genetically related to monkey B virus and human herpes simplex virus (HSV). To study the temporal progression of viral spread and associated lesions, Balb/c mice were inoculated epidermally by scarification with a green fluorescent protein (GFP)-expressing recombinant strain of SaHV-1 and killed sequentially. Pinpoint ulcerative lesions in the inoculated epidermis progressed over a few days to unilateral or bilateral hindlimb paresis or paralysis, urinary and faecal incontinence, abdominal distension, hunched posture and eventual depression warranting euthanasia. Viral replication was present within epidermal keratinocytes, neurons of the dorsal root ganglia and thoracolumbar spinal cord, regional autonomic ganglia, lower urinary tract epithelium and colonic myenteric plexuses, as indicated by histological lesions and GFP expression. Almost all mice inoculated with 10(5) or 10(6) plaque-forming units (PFU) of SaHV-1 developed rapidly progressive disease. Two of eight mice given 10(4)PFU developed disease, but no mice receiving less than 10(4)PFU gave evidence of infection. Mice that showed no clinical signs also failed to develop an antiviral IgG response, indicating absence of active viral infection. For SaHV-1 inoculated epidermally, the ID(50), CNSD(50) and LD(50) values were identical (10(4.38)), indicating that successful infection by this route invariably resulted in lethal CNS (central nervous system) disease. Consistently severe disease in all infected animals, with regionally extensive distribution of viral replication, constituted a marked difference from the disease produced by intramuscular inoculation.
Collapse
Affiliation(s)
- M A Breshears
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | |
Collapse
|
22
|
Scanlan PM, Tiwari V, Bommireddy S, Shukla D. Spinoculation of heparan sulfate deficient cells enhances HSV-1 entry, but does not abolish the need for essential glycoproteins in viral fusion. J Virol Methods 2005; 128:104-12. [PMID: 15908019 DOI: 10.1016/j.jviromet.2005.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Revised: 04/04/2005] [Accepted: 04/05/2005] [Indexed: 10/25/2022]
Abstract
Cell surface heparan sulfate functions as a co-receptor in HSV-1 entry. In order to study its significance in context with specific gD receptors (nectin-1, HVEM, and 3-O-sulfated heparan sulfate) a low speed centrifugation based virus inoculation (spinoculation) method was used. The experiments were performed at 1200 x g using glycosylaminoglycan positive (GAG+) or deficient (GAG-) cells expressing gD receptors. Clearly, spinoculation of GAG- nectin-1 or HVEM cells enhanced significantly viral entry compared to similar but unspun cells. The enhanced entry was due to increased virus deposition at the cell surface and not due to pelleting of the virus. Among the gD receptors, spinoculated GAG- HVEM cells showed restoration of HSV-1 entry compared to unspinoculated GAG+ HVEM cells. In contrast, spinoculated GAG- nectin-1 cells showed less entry than unspinoculated GAG+ nectin-1 cells. GAG- 3-O-sulfotransferase-expressing cells or heparinase treated GAG+ 3-O-sulfated heparan sulfate cells, in contrast, remained resistant to entry even after spinoculation. To investigate further, any potential effects of centrifugation on membrane fusion, a virus-free cell fusion assay was performed. Clearly, spinning had no effects on cell fusion, nor could it replace the need for all four essential glycoproteins. Taken together these results suggest that heparan sulfate plays a role of an attachment receptor, which could be substituted by spinoculation. This effect, however, varies with the gD receptor used, which in turn, could be used as a means for identifying gD receptor usage for entry into a cell type.
Collapse
Affiliation(s)
- Perry M Scanlan
- Department of Microbiology-Immunology, College of Medicine, University of Illinois at Chicago, Lions of Illinois Eye Research Institute, M/C 648, 1855 West Taylor Street, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
23
|
Ellermann-Eriksen S. Macrophages and cytokines in the early defence against herpes simplex virus. Virol J 2005; 2:59. [PMID: 16076403 PMCID: PMC1215526 DOI: 10.1186/1743-422x-2-59] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 08/03/2005] [Indexed: 11/12/2022] Open
Abstract
Herpes simplex virus (HSV) type 1 and 2 are old viruses, with a history of evolution shared with humans. Thus, it is generally well-adapted viruses, infecting many of us without doing much harm, and with the capacity to hide in our neurons for life. In rare situations, however, the primary infection becomes generalized or involves the brain. Normally, the primary HSV infection is asymptomatic, and a crucial element in the early restriction of virus replication and thus avoidance of symptoms from the infection is the concerted action of different arms of the innate immune response. An early and light struggle inhibiting some HSV replication will spare the host from the real war against huge amounts of virus later in infection. As far as such a war will jeopardize the life of the host, it will be in both interests, including the virus, to settle the conflict amicably. Some important weapons of the unspecific defence and the early strikes and beginning battle during the first days of a HSV infection are discussed in this review. Generally, macrophages are orchestrating a multitude of anti-herpetic actions during the first hours of the attack. In a first wave of responses, cytokines, primarily type I interferons (IFN) and tumour necrosis factor are produced and exert a direct antiviral effect and activate the macrophages themselves. In the next wave, interleukin (IL)-12 together with the above and other cytokines induce production of IFN-gamma in mainly NK cells. Many positive feed-back mechanisms and synergistic interactions intensify these systems and give rise to heavy antiviral weapons such as reactive oxygen species and nitric oxide. This results in the generation of an alliance against the viral enemy. However, these heavy weapons have to be controlled to avoid too much harm to the host. By IL-4 and others, these reactions are hampered, but they are still allowed in foci of HSV replication, thus focusing the activity to only relevant sites. So, no hero does it alone. Rather, an alliance of cytokines, macrophages and other cells seems to play a central role. Implications of this for future treatment modalities are shortly considered.
Collapse
Affiliation(s)
- Svend Ellermann-Eriksen
- Department of Clinical Microbiology, Aarhus University Hospital, Skejby Sygehus, Brendstrupgaardsvej 100, DK-8200 Aarhus N., Denmark.
| |
Collapse
|
24
|
Waddington SN, Kennea NL, Buckley SMK, Gregory LG, Themis M, Coutelle C. Fetal and neonatal gene therapy: benefits and pitfalls. Gene Ther 2004; 11 Suppl 1:S92-7. [PMID: 15454963 DOI: 10.1038/sj.gt.3302375] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The current approaches to gene therapy of monogenetic diseases into mature organisms are confronted with several problems including the following: (1) the underlying genetic defect may have already caused irreversible pathological changes; (2) the level of sufficient protein expression to ameliorate or prevent the disease requires prohibitively large amounts of gene delivery vector; (3) adult tissues may be poorly infected by conventional vector systems dependent upon cellular proliferation for optimal infection, for example, oncoretrovirus vectors; (4) immune responses, either pre-existing or developing following vector delivery, may rapidly eliminate transgenic protein expression and prevent future effective intervention. Early gene transfer, in the neonatal or even fetal period, may overcome some or all of these obstacles. The mammalian fetus enjoys a uniquely protected environment in the womb, bathed in a biochemically and physically supportive fluid devoid of myriad extra-uterine pathogens. Strong physical and chemical barriers to infection might, perhaps, impede the frenetic cell division. The physical support and the biochemical support provided by the fetal-maternal placental interface may, therefore, minimize the onset of genetic diseases manifest early in life. The fetal organism must prepare itself for birth, but lacking a mature adaptive immune system may depend upon more primordial immune defences. It is the nature of these defences, and the vulnerabilities they protect, that are poorly understood in the context of gene therapy and might provide useful information for approaches to gene therapy in the young, as well as perhaps the mature organism.
Collapse
Affiliation(s)
- S N Waddington
- Imperial College London, Gene Therapy Research Group, Section of Cell and Molecular Biology, Division of Biomedical Sciences, Sir Alexander Fleming Building, Imperial College Road, London, UK
| | | | | | | | | | | |
Collapse
|
25
|
Li ZY, Ni S, Yang X, Kiviat N, Lieber A. Xenograft models for liver metastasis: Relationship between tumor morphology and adenovirus vector transduction. Mol Ther 2004; 9:650-7. [PMID: 15120325 DOI: 10.1016/j.ymthe.2004.01.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Accepted: 01/30/2004] [Indexed: 12/28/2022] Open
Abstract
The improvement of initial tumor cell transduction with viral vectors is a major task in tumor gene therapy. We have developed mouse tumor models with hepatic metastases to study transduction of tumor cells after systemic adenovirus vector application. The tumor models were established by intraportal transplantation of human tumor cell lines into immunodeficient mice. Liver metastases derived from cervix, colon, breast, and liver cancer lines were analyzed for distribution of extracellular matrix, vascularization, and transgene expression after tail vein injection of adenovirus vectors. Overall, xenografts resembled the morphology of corresponding tumors in cancer patients. Adenovirus-mediated gene delivery depended on tumor vascularization and direct contact between blood vessels and tumor cells. These models represent important tools for studying and improving tumor gene therapy approaches.
Collapse
Affiliation(s)
- Zong-Yi Li
- Division of Medical Genetics, Department of Medicine, University of Washington, Box 357720, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
26
|
Karpati G. Molecular therapies for the nervous system and muscle. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2004:159-78. [PMID: 12894456 DOI: 10.1007/978-3-662-05352-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Affiliation(s)
- G Karpati
- Montreal Neurological Institute and Hospital, 3801 rue University Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
27
|
Li J, Wang D, Qian S, Chen Z, Zhu T, Xiao X. Efficient and long-term intracardiac gene transfer in delta-sarcoglycan-deficiency hamster by adeno-associated virus-2 vectors. Gene Ther 2003; 10:1807-13. [PMID: 12960970 DOI: 10.1038/sj.gt.3302078] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intracardiac gene transfer and gene therapy have been investigated with different vector systems. Here we used adeno-associated virus (AAV) vectors to deliver either a reporter gene or a therapeutic gene into the heart of golden Syrian hamsters. The method of gene delivery was direct infusion of the AAV2 vectors into the coronary artery ex vivo in a heterotopically transplanted heart. When an AAV2 vector carrying the Lac-Z gene driven by CMV promoter was delivered into the heart of healthy hamsters, effective gene transfer was achieved in up to 90% of the cardiomyocytes. Lac-Z gene expression persisted for more than 1 year without immune rejection or promoter shutoff. Furthermore, when an AAV2 vector carrying human delta-sarcoglycan gene was similarly delivered into the heart of Bio14.6 Syrian hamster, a congestive heart failure and limb girdle muscular dystrophy animal model, widespread therapeutic gene transfer was achieved in a majority of the cardiomyocytes. Efficient expression of the human delta-sarcoglycan gene in the dystrophic hamster hearts restored the entire sarcoglycan complex that was missing due to the primary deficiency of delta-sarcoglycan. Transgene expression persisted for 4 months (the duration of the study) without immune rejection or promoter shutoff. These results indicate that AAV is a promising vector system for cardiac gene therapy.
Collapse
Affiliation(s)
- J Li
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
28
|
Fu X, Meng F, Tao L, Jin A, Zhang X. A strict-late viral promoter is a strong tumor-specific promoter in the context of an oncolytic herpes simplex virus. Gene Ther 2003; 10:1458-64. [PMID: 12900760 DOI: 10.1038/sj.gt.3302029] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Confinement of transgene expression to target cells is highly desirable in gene therapy. Current strategies of transcriptional targeting to tumors usually rely on tissue-specific promoters to control gene expression. However, such promoters generally have much lower activity than the constitutive viral promoters. We have explored an alternative approach, using a strict-late viral promoter (UL38p) in the context of an oncolytic herpes simplex virus (HSV) for tumor-selective gene expression. As with many DNA viruses, the genomic transcription of HSV is a tightly regulated molecular cascade in which early and late phases of gene expression are separated by viral DNA replication. In particular, some of the late transcripts are categorized as strict-late, whose expression depends rigorously on the initiation of viral DNA replication. Our in vitro and in vivo characterization showed that in normal nondividing cells, where the oncolytic HSV has limited ability to replicate, the UL38p has minimal activity. However, in tumor or cycling cells where the virus can fully replicate, transgene expression from UL38p was almost as high as from the cytomegalovirus immediate-early promoter. These results suggest that delivery of therapeutic genes driven by UL38p through an oncolytic HSV may be an effective approach to gene therapy for malignant diseases.
Collapse
Affiliation(s)
- X Fu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
29
|
Curi MA, Skelly CL, Meyerson SL, Baldwin ZK, Balasubramanian V, Advani SJ, Glagov S, Roizman B, Weichselbaum RR, Schwartz LB. Sustained inhibition of experimental neointimal hyperplasia with a genetically modified herpes simplex virus. J Vasc Surg 2003; 37:1294-300. [PMID: 12764278 DOI: 10.1016/s0741-5214(02)75333-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Reported herein is a potential strategy for sustained smooth muscle cell (SMC) inhibition with a virulence-attenuated herpes simplex virus (HSV). Experiments were conducted in vitro to demonstrate selective SMC cytotoxicity and in vivo to demonstrate reduced neointimal hyperplasia (NIH) in a clinically relevant animal model. METHODS In vitro: Cultured human umbilical artery smooth muscle cells (UASMC) and venous endothelial cells (HUVEC) were exposed to varying multiplicities of infection (MOI) of a gamma(1)34.5-deleted HSV-1 virus (R849). Cell survival was assessed at 48 and 72 hours with a colorimetric MTT viability assay. In vivo: New Zealand White rabbit external jugular veins (n = 21) were exposed to R849 (2.5 x 10(6) pfu/mL) or culture medium at 110 to 120 mm Hg for 10 minutes, then fashioned as vein patches on carotid arteries. Carotid arteries were ligated distally to decrease blood flow and stimulate a hyperplastic response (ultra-low shear stress model). After 2, 4, 12, and 24 weeks, patched segments were perfusion-fixed with glutaraldehyde and morphometrically examined for NIH formation. RESULTS In vitro: At 48 hours, R849 exhibited preferential cytotoxicity to UASMC compared with HUVEC, with 11% +/- 10% of UASMCs and 49% +/- 8% of HUVECs surviving after infection with MOI = 25 (P <.05). Higher MOI resulted in poor survival of both cell lines. In vivo: Blood flow was similarly reduced in all animals both at surgery (0.9 +/- 0.1 mL/min vs 1.6 +/- 0.3 mL/min) and at harvest (2.7 +/- 0.4 mL/min vs 2.5 +/- 0.5 mL/min). R849-infected patches exhibited markedly less NIH than control patches did at 2 weeks (162 +/- 14 microm vs 49 +/- 6 microm; P <.05), 4 weeks (190 +/- 27 microm vs 67 +/- 8 microm; P <.05), and 12 weeks (233 +/- 18 microm vs 113 +/- 2 microm; P <.05). CONCLUSION The virulence-attenuated HSV strain R849 demonstrates selective cytotoxicity for SMC and is capable of sustained inhibition of NIH in an experimental model of vein graft failure.
Collapse
Affiliation(s)
- Michael A Curi
- Section of Vascular Surgery, Department of Surgery, University of Chicago MC 5028, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The development of therapeutic strategies that overcome the unique problems posed by Duchenne muscular dystrophy (DMD) has lead to the development of many contemporary approaches to human disease in general. Various treatment approaches have been explored--such as pharmacological therapies and cell-based, cytokine, and genetic therapies--that are all targeted to specific features of dystrophic DMD muscle pathology. In genetic therapies, the large size of the dystrophin gene has necessitated the development and use of novel functional minidystrophin and microdystrophin genes, muscle-specific promoter systems, and gutted adenoviral systems. In addition to these well defined viral strategies, plasmid vectors and the upregulation of utrophin (a dystrophin homologue) have potential. Various novel genetic approaches--such as antisense-mediated exon skipping, gene correction, and new cytokine approaches--are also being developed. Together these exciting developments bring an effective treatment for DMD closer than ever before.
Collapse
|
31
|
Abstract
Herpes simplex virus (HSV) is a neurotropic DNA virus with many favorable properties as a gene delivery vector. HSV is highly infectious, so HSV vectors are efficient vehicles for the delivery of exogenous genetic material to cells. Viral replication is readily disrupted by null mutations in immediate early genes that in vitro can be complemented in trans, enabling straightforward production of high-titre pure preparations of non-pathogenic vector. The genome is large (152 Kb) and many of the viral genes are dispensable for replication in vitro, allowing their replacement with large or multiple transgenes. Latent infection with wild-type virus results in episomal viral persistence in sensory neuronal nuclei for the duration of the host lifetime. Transduction with replication-defective vectors causes a latent-like infection in both neural and non-neural tissue; the vectors are non-pathogenic, unable to reactivate and persist long-term. The latency active promoter complex can be exploited in vector design to achieve long-term stable transgene expression in the nervous system. HSV vectors transduce a broad range of tissues because of the wide expression pattern of the cellular receptors recognized by the virus. Increasing understanding of the processes involved in cellular entry has allowed preliminary steps to be taken towards targeting the tropism of HSV vectors. Using replication-defective HSV vectors, highly encouraging results have emerged from recent pre-clinical studies on models of neurological disease, including glioma, peripheral neuropathy, chronic pain and neurodegeneration. Consequently, HSV vectors encoding appropriate transgenes to tackle these pathogenic processes are poised to enter clinical trials.
Collapse
Affiliation(s)
- Edward A Burton
- Department of Clinical Neurology, University of Oxford, United Kingdom
| | | | | |
Collapse
|
32
|
Thirion C, Larochelle N, Volpers C, Dunant P, Stucka R, Holland P, Nalbantoglu J, Kochanek S, Lochmüller H. Strategies for muscle-specific targeting of adenoviral gene transfer vectors. Neuromuscul Disord 2002; 12 Suppl 1:S30-9. [PMID: 12206792 DOI: 10.1016/s0960-8966(02)00079-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Currently, adenoviral transfer of therapeutic genes such as dystrophin is hampered by low transduction efficiency of adult skeletal muscle. This is largely due to the lack of appropriate virus attachment receptors on the myofiber surface. Recent studies in transgenic mice revealed that upregulation of Coxsackie- and adenovirus receptor improves gene transfer efficiency by approximately ten-fold. Conversely, the vector load that needed to be administered to achieve sufficient gene transfer could be lowered significantly. Reduced viral vector loads may help to control virally mediated toxicity and immunogenicity. To date, there are no drugs or methods known to increase Coxsackie- and adenovirus receptor expression in skeletal muscle that would be easily applicable in humans. However, alternative strategies such as vector retargeting are currently being investigated that may allow for an increase in binding of adenoviral vectors to skeletal muscle. Recent experiments have shown that directed mutagenesis of the adenoviral fiber knob allows for a significant reduction in Coxsackie- and adenovirus receptor binding and for introduction of a new binding domain. Therefore, vector retargeting towards efficient and specific infection of skeletal muscle may be achieved by directed genetic alteration of adenoviral capsid proteins.
Collapse
|
33
|
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
|
34
|
Gournay J, Auvigne I, Pichard V, Ligeza C, Bralet MP, Ferry N. In vivo cell lineage analysis during chemical hepatocarcinogenesis in rats using retroviral-mediated gene transfer: evidence for dedifferentiation of mature hepatocytes. J Transl Med 2002; 82:781-8. [PMID: 12065689 DOI: 10.1097/01.lab.0000017363.11489.ad] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Feeding adult rats with a diet containing 2-acetylaminofluorene (2-AAF) results in suppression of hepatocyte proliferation and stimulation of oval cell proliferation. Although oval cells may be facultative liver stem cells, the actual relationship between oval cells and liver cancer has not been clearly established in vivo. Our goal was to label hepatic cells in vivo using retroviral vectors and follow their fate during the early steps of chemically induced hepatocarcinogenesis. Oval cell proliferation was induced by continuous feeding with a carcinogenic diet containing 2-AAF. We used two different strategies to genetically label hepatic cells: (a) labeling of proliferating cells in rats fed 2-AAF by injecting recombinant retroviral vectors containing the beta-galactosidase gene either in a peripheral vein or in the common bile duct at the peak of oval cell proliferation and (b) prelabeling of hepatocytes by intravenously injecting recombinant vectors 1 day after partial hepatectomy and 1 week before subsequent administration of 2-AAF. Using the first strategy, transgene expression occurred in both oval cells and hepatocytes. Using the second strategy, we could selectively label, and hence study the fate of, differentiated hepatocytes. In the latter case, we observed clusters of beta-galactosidase-positive hepatocytes, some of them also expressing preneoplastic markers such as gamma-glutamyl transpeptidase as well as the placental form of glutathione-S-transferase. These results demonstrate that preneoplastic foci can originate from mature hepatocytes and are consistent with the hypothesis that dedifferentiation of mature hepatocytes may occur during the course of carcinogenic regimen.
Collapse
Affiliation(s)
- Jérôme Gournay
- Laboratoire de Thérapie Génique, Hôtel-Dieu, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | | | | | | | | | | |
Collapse
|
35
|
Wang Y, Mukherjee S, Fraefel C, Breakefield XO, Allen PD. Herpes simplex virus type 1 amplicon vector-mediated gene transfer to muscle. Hum Gene Ther 2002; 13:261-73. [PMID: 11812282 DOI: 10.1089/10430340252769789] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) amplicon vectors were evaluated for feasibility in gene therapy of Duchenne's muscular dystrophy (DMD). An amplicon vector expressing enhanced green fluorescent protein (eGFP) was examined for transduction efficiency and cytotoxicity in cultured muscle cells, and for transduction efficiency, duration of transgene expression, and immunogenicity in tibialis anterior (TA) muscles of neonatal mice. Transduction efficiencies in murine and human myoblasts were 60-90 and 50-60%, respectively, when myoblasts were transduced at multiplicities of infection (MOIs) of 1-5. Similar transduction efficiencies were observed in myotubes of both species. No cytotoxic effects were noticed at an MOI of 10, the highest MOI tested. An amplicon vector, HyMD, containing the full-length mouse dystrophin cDNA and its muscle creatine kinase (MCK) promoter-enhancer, with a total size of 26 kb, was constructed and used to transduce mdx mouse myotubes. The expression of dystrophin in these cells was demonstrated by immunocytochemistry. After injecting 4-6 x 10(5) transduction units (TU) of HSVGN amplicon vectors, 10-50% of myofibers in the injected TA muscles expressed GFP. Although transgene expression was attenuated over time, significant improvement in long-term transgene expression and persistence of vector DNA was achieved, when compared with the first generation of recombinant HSV-1 vectors. Immunohistochemistry showed a modest CD4(+) lymphocyte infiltration in the vicinity of the injection. A gradually developed CD8(+) lymphocyte infiltration was also seen, most likely related to the antigenicity of the transgene product, GFP. We conclude that the HSV-1 amplicon vector is a promising vehicle for gene delivery in DMD. However, new strategies need to be evaluated to increase the stability of transgene expression.
Collapse
Affiliation(s)
- Yaming Wang
- Department of Anesthesia, Brigham and Women's Hospital, Massachusetts General Hospital, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
36
|
Burton EA, Wechuck JB, Wendell SK, Goins WF, Fink DJ, Glorioso JC. Multiple applications for replication-defective herpes simplex virus vectors. Stem Cells 2002; 19:358-77. [PMID: 11553845 DOI: 10.1634/stemcells.19-5-358] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Herpes simplex virus (HSV) is a neurotropic DNA virus. The viral genome is large (152 kb), and many genes are dispensable for viral function, allowing insertion of multiple or large transgene expression cassettes. The virus life cycle includes a latent phase, during which the viral genome remains as a stable episomal element within neuronal nuclei for the lifetime of the host, without disturbing normal function. We have exploited these features of HSV to construct a series of nonpathogenic gene therapy vectors that efficiently deliver therapeutic and experimental transgenes to neural and non-neural tissue. Importantly, transgene expression may be sustained long term; reporter gene expression has been demonstrated for over a year in the nervous system. This article discusses the generation of replication-defective HSV vectors and reviews recent studies investigating their use in several animal models of human disease. We have demonstrated correction or prevention of a number of important neurological phenotypes, including neurodegeneration, chronic pain, peripheral neuropathy, and malignancy. In addition, HSV-mediated transduction of non-neurological tissues allows their use as depot sites for synthesis of circulating and locally acting secreted proteins. New applications for this vector system include the genetic modification of stem cell populations; this may become an important means to direct cellular differentiation or deliver therapeutic genes systemically. Replication-defective HSV vectors are an effective and flexible vehicle for the delivery of transgenes to numerous tissues, with multiple applications.
Collapse
Affiliation(s)
- E A Burton
- University of Pittsburgh School of Medicine, Department of Molecular Genetics and Biochemistry, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | | | |
Collapse
|
37
|
Shukla D, Spear PG. Herpesviruses and heparan sulfate: an intimate relationship in aid of viral entry. J Clin Invest 2001. [DOI: 10.1172/jci200113799] [Citation(s) in RCA: 361] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
38
|
Shukla D, Spear PG. Herpesviruses and heparan sulfate: an intimate relationship in aid of viral entry. J Clin Invest 2001; 108:503-10. [PMID: 11518721 PMCID: PMC209412 DOI: 10.1172/jci13799] [Citation(s) in RCA: 267] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- D Shukla
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | |
Collapse
|
39
|
Rissanen TT, Vajanto I, Ylä-Herttuala S. Gene therapy for therapeutic angiogenesis in critically ischaemic lower limb - on the way to the clinic. Eur J Clin Invest 2001; 31:651-66. [PMID: 11473566 DOI: 10.1046/j.1365-2362.2001.00864.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Currently, no effective pharmacological treatment is available for vascularisation defects in lower limbs. Many patients presenting with persistent pain and ischaemic ulcers are not suitable candidates for surgical or endovascular approaches. Further refinement of the available methods will undoubtedly lead to a more active approach towards treatment of peripheral arterial occlusive disease (PAOD). Recently, therapeutic angiogenesis, in the form of recombinant growth factor administration or gene therapy, has emerged as a novel tool to treat these patients. However, improved gene transfer methods and better understanding of blood vessel formation are required to bring therapeutic angiogenesis to clinical practice. Here we review the clinical problem (PAOD), mechanisms of blood vessel formation (angiogenesis, vasculogenesis and arteriogenesis), experimental evidence and clinical trials for therapeutic angiogenesis in critically ischaemic lower limbs. Also, angiogenic growth factors, including vascular endothelial growth factors (VEGFs) and fibroblast growth factors (FGFs), delivery methods, and vectors for gene transfer in skeletal muscle, are discussed. In addition to vascular growth, gene transfer of growth factors may enhance regeneration, survival, and innervation of ischaemic skeletal muscle. Nitric oxide (NO) appears to be a key mediator in vascular homeostasis and growth, and a reduction in its production by age, hypercholesterolemia or diabetes leads to the impairment of ischaemic disorders.
Collapse
Affiliation(s)
- T T Rissanen
- A. I. Virtanen Institute, Department of Medicine, University of Kuopio, Kuopio, Finland
| | | | | |
Collapse
|
40
|
Rahkila P, Takala TE, Parton RG, Metsikkö K. Protein targeting to the plasma membrane of adult skeletal muscle fiber: an organized mosaic of functional domains. Exp Cell Res 2001; 267:61-72. [PMID: 11412038 DOI: 10.1006/excr.2001.5101] [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: 11/22/2022]
Abstract
The plasma membrane of differentiated skeletal muscle fibers comprises the sarcolemma, the transverse (T) tubule network, and the neuromuscular and muscle-tendon junctions. We analyzed the organization of these domains in relation to defined surface markers, beta-dystroglycan, dystrophin, and caveolin-3. These markers were shown to exhibit highly organized arrays along the length of the fiber. Caveolin-3 and beta-dystroglycan/dystrophin showed distinct, but to some extent overlapping, labeling patterns and both markers left transverse tubule openings clear. This labeling pattern revealed microdomains over the entire plasma membrane with the exception of the neuromuscular and muscle-tendon junctions which formed distinct demarcated macrodomains. Our results suggest that the entire plasma membrane of mature muscle comprises a mosaic of T tubule domains together with sareolemmal caveolae and beta-dystroglycan domains. The domains identified with these markers were examined with respect to targeting of viral proteins and other expresseddomain-specific markers. We found that each marker protein was targeted to distinct microdomains. The macrodomains were intensely labeled with all our markers. Replacing the cytoplasmic tail of the vesicular stomatitis virus glycoprotein with that of CD4 resulted in retargeting from one domain to another. The domain-specific protein distribution at the muscle cell surface may be generated by targeting pathways requiring specific sorting information but this trafficking is different from the conventional apical-basolateral division.
Collapse
Affiliation(s)
- P Rahkila
- Department of Anatomy and Cell Biology, University of Oulu, Oulu, 90014, Finland.
| | | | | | | |
Collapse
|
41
|
Cao B, Pruchnic R, Ikezawa M, Xiao X, Li J, Wickham TJ, Kovesdi I, Rudert WA, Huard J. The role of receptors in the maturation-dependent adenoviral transduction of myofibers. Gene Ther 2001; 8:627-37. [PMID: 11320409 DOI: 10.1038/sj.gt.3301425] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2000] [Accepted: 01/15/2001] [Indexed: 11/09/2022]
Abstract
One of the major hurdles facing the application of adenoviral gene transfer to skeletal muscle is the maturation-dependent transduction of muscle myofibers. It was recently proposed that the viral receptors (Coxsackie and adenovirus receptor (CAR) and the integrins alphavbeta3/beta5) play a major role in the poor adenoviral transduction of mature myofibers. Here we report the findings of morphological studies designed to determine experimentally the role of receptors in the adenoviral transduction of mature myofibers. First, we observed that the expression of both attachment and internalization receptors did not change significantly during muscle development. Second, when an extended tropism adenoviral vector (AdPK) that attaches to heparan sulfate proteoglycan (HSP) is used, a significant reduction of adenoviral transduction still occurs in mature myofibers despite HSP's high expression in mature skeletal muscle fibers. Third, when the adeno-associated virus (AAV) is used, which also utilizes HSP as a viral receptor, muscle fibers at different maturities can be highly transduced. Fourth, the pre-irradiation of the skeletal muscle of newborn mice to inactivate myoblasts dramatically decreased the transduction level of Ad and AdPK, but had no effect on AAV-mediated viral transduction of immature myofibers. These results taken together suggest that the viral receptor(s) is not a major determinant in maturation-dependent adenoviral transduction of myofibers.
Collapse
Affiliation(s)
- B Cao
- Growth and Development Laboratory, Department of Orthopaedic Surgery, Children's Hospital of Pittsburgh and University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Petersen ZQ, Huard J. The influence of muscle fiber type in myoblast-mediated gene transfer to skeletal muscles. Cell Transplant 2000; 9:503-17. [PMID: 11038067 DOI: 10.1177/096368970000900407] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Myoblast transplantation has been hindered by immune rejection problems, as well as the poor survival and spread of transplanted cells. Our recent study has shown that the poor survival of the injected cells can be totally overcome by the use of specific populations of muscle-derived cells. In the present study, we have investigated whether a relationship exists between the fate of transplanted cells and the muscle fiber types. Four kinds of myogenic cells [primary myoblasts at a high purity (PMb), myoblasts isolated from fast single fibers (FMb), mdx (MCL), and MtMd-1 cell lines] were infected with an adenoviral vector carrying a LacZ reporter gene and injected into mdx hindlimb muscle. The LacZ transduced myofibers formed by the fusion of the injected myoblasts at 2-10 days postinjection were colocalized with MyHC stainings. The PMb cells, which expressed both slow and fast MyHCs in vitro, displayed the same phenotypes when injected into the m. soleus and m. gastrocnemius (white) muscles, which contained 70% and 0% of slow myofibers, respectively, and showed a high degree of fusion with host muscle fibers. In contrast, the FMb cells only expressed fast MyHCs in vitro and fused exclusively with each other or with host fast muscle fibers when injected in the m. gastrocnemius. Injected MCL and MtMd-1 fused predominantly with each other and displayed a similar expression of MyHCs to those they expressed in vitro. Just a few host myofibers were found to express the reporter gene product following implantation of both cell lines, indicating that these myogenic cell lines display an intrinsic potential to fuse together rather than with host myofibers. Based on the data, we concluded that 1) the essential key to survival is the ability of the donor cells to fuse with the host myofibers, and 2) the most successful combination is achieved between donor primary muscle cells that express both fast and slow MyHC and a host muscle type that facilitates fusion.
Collapse
Affiliation(s)
- Z Q Petersen
- Department of Orthopaedic Surgery and Molecular Genetics & Biochemistry, Musculoskeletal Research Center, Children's Hospital of Pittsburgh and University of Pittsburgh, PA 15261, USA
| | | |
Collapse
|
43
|
Bartlett RJ, Stockinger S, Denis MM, Bartlett WT, Inverardi L, Le TT, thi Man N, Morris GE, Bogan DJ, Metcalf-Bogan J, Kornegay JN. In vivo targeted repair of a point mutation in the canine dystrophin gene by a chimeric RNA/DNA oligonucleotide. Nat Biotechnol 2000; 18:615-22. [PMID: 10835598 DOI: 10.1038/76448] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the canine model of Duchenne muscular dystrophy in golden retrievers (GRMD), a point mutation within the splice acceptor site of intron 6 leads to deletion of exon 7 from the dystrophin mRNA, and the consequent frameshift causes early termination of translation. We have designed a DNA and RNA chimeric oligonucleotide to induce host cell mismatch repair mechanisms and correct the chromosomal mutation to wild type. Direct skeletal muscle injection of the chimeric oligonucleotide into the cranial tibialis compartment of a six-week-old affected male dog, and subsequent analysis of biopsy and necropsy samples, demonstrated in vivo repair of the GRMD mutation that was sustained for 48 weeks. Reverse transcription-polymerase chain reaction (RT-PCR) analysis of exons 5-10 demonstrated increasing levels of exon 7 inclusion with time. An isolated exon 7-specific dystrophin antibody confirmed synthesis of normal-sized dystrophin product and positive localization to the sarcolemma. Chromosomal repair in muscle tissue was confirmed by restriction fragment length polymorphism (RFLP)-PCR and sequencing the PCR product. This work provides evidence for the long-term repair of a specific dystrophin point mutation in muscle of a live animal using a chimeric oligonucleotide.
Collapse
Affiliation(s)
- R J Bartlett
- Department of Veterinary Medicine and Surgery, Dalton Cardiovascular Research Center, College of Veterinary Medicine, Columbia, MO 65202, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Logeart D, Hatem SN, Rücker-Martin C, Chossat N, Névo N, Haddada H, Heimburger M, Perricaudet M, Mercadier JJ. Highly efficient adenovirus-mediated gene transfer to cardiac myocytes after single-pass coronary delivery. Hum Gene Ther 2000; 11:1015-22. [PMID: 10811230 DOI: 10.1089/10430340050015329] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Efficient and homogeneous gene transfer to cardiac myocytes is a major target in myocardial gene therapy. The aim of this study was to determine the conditions permitting efficient, homogeneous, adenovirus-mediated gene transfer to cardiac myocytes, with a view to application during coronary artery catheterization. Gene transfer to adult rat ventricular myocytes was conducted using type 5 adenoviruses carrying the lacZ reporter gene. Adenovirus delivery via coronary arteries was performed on isolated perfused rat hearts, and gene transfer efficiency was analyzed on whole ventricles, freshly isolated myocytes, and cultured myocytes. Single-pass delivery of 1 X 10(9) PFU associated with 1 min of no-flow yielded only 1 +/- 0.5% of positive myocytes. Pretreatment by histamine perfusion (10(-5) M final concentration) increased this value to 30 +/- 9% (p < 0.001), and pretreatment by Ca2+-free buffer perfusion increased it to 67 +/- 8% (p < 0.001). Combination of the two pretreatments had no additional effect. Increasing the viral dose to 3 X 10(9) PFU increased transfection efficiency only in permeabilized vessels. The 1-min no-flow period after adenovirus delivery was crucial for efficient gene transfer: despite histamine pretreatment, only 2 +/- 1% positive myocytes were observed without flow interruption (p < 0.05 versus 1 min of no-flow). Gene transfer was shown to occur in situ during cardiac perfusion, rather than during heart digestion or myocyte isolation. This study shows that highly efficient adenovirus-mediated gene transfer to cardiac myocytes in situ can be achieved by single-pass intracoronary vector delivery, provided that vascular permeability is first increased and coronary flow is briefly interrupted.
Collapse
Affiliation(s)
- D Logeart
- INSERM U 460, Faculté de Médecine Xavier Bichat, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pruchnic R, Cao B, Peterson ZQ, Xiao X, Li J, Samulski RJ, Epperly M, Huard J. The use of adeno-associated virus to circumvent the maturation-dependent viral transduction of muscle fibers. Hum Gene Ther 2000; 11:521-36. [PMID: 10724031 DOI: 10.1089/10430340050015716] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Muscle-based gene therapy using adenovirus, retrovirus, and herpes simplex virus has been hindered by viral cytotoxicity, host immune response, and the maturation-dependent viral transduction of muscle fibers. The development of new mutant vectors has greatly reduced the toxicity and the immune rejection problems, but the inability of viral vectors to penetrate and transduce mature myofibers remains an important issue. Research has been focused on the characterization of barriers to viral transduction in mature myofibers to develop strategies to circumvent the maturation-dependent viral transduction of myofibers. Here, we report that adeno-associated virus (AAV) can be used to overcome the maturation-dependent viral transduction of myofibers. We have investigated by which mechanism AAV can penetrate and efficiently transduce mature muscle fibers, and have shown that this viral vector is not blocked by the basal lamina and that AAV transduction of myofibers is independent of myoblast mediation. Although AAV can efficiently transduce mature myofibers, a differential transduction is still observed among the different types of myofibers that correlates with the expression of the heparan sulfate proteoglycan receptors, the muscle maturity, the number of viral particles used, and the time postinjection. The identification of the mechanisms by which AAV transduces mature myofibers will help in the development of strategies to achieve an efficient muscle-based gene therapy for inherited and acquired diseases.
Collapse
Affiliation(s)
- R Pruchnic
- Department of Orthopedic Surgery, Children's Hospital of Pittsburgh and University of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kasemkijwattana C, Menetrey J, Bosch P, Somogyi G, Moreland MS, Fu FH, Buranapanitkit B, Watkins SS, Huard J. Use of growth factors to improve muscle healing after strain injury. Clin Orthop Relat Res 2000:272-85. [PMID: 10660723 DOI: 10.1097/00003086-200001000-00028] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Muscle injuries represent a large number of professional and recreational sports injuries. Muscle strains habitually occur after an eccentric contraction, which often leads to an injury located in the myotendinous junction. Treatment varies widely, depending on the severity of the trauma, but has remained limited mostly to rest, ice, compression, elevation, antiinflammatory drugs, and mobilization. The authors' research group aims to develop new biologic approaches to improve muscle healing after injuries, including muscle strains. To achieve this goal, the authors investigated several parameters that will lead to the development of new strategies to enhance muscle healing. The authors first evaluated natural muscle healing after strain injuries and showed that muscle regeneration occurs in the early phase of healing but becomes impaired with time by the development of tissue fibrosis. Several growth factors capable of improving muscle regeneration were investigated; basic fibroblast growth factor, insulin-like growth factor, and nerve growth factors were identified as substances capable of enhancing muscle regeneration and improving muscle force in the strained injured muscle. The current study should aid in the development of strategies to promote efficient muscle healing and complete recovery after strain injury.
Collapse
Affiliation(s)
- C Kasemkijwattana
- Department of Orthopaedic Surgery, University of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
van Deutekom JC, Cao B, Pruchnic R, Wickham TJ, Kovesdi I, Huard J. Extended tropism of an adenoviral vector does not circumvent the maturation-dependent transducibility of mouse skeletal muscle. J Gene Med 1999; 1:393-9. [PMID: 10753064 DOI: 10.1002/(sici)1521-2254(199911/12)1:6<393::aid-jgm65>3.0.co;2-u] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Efficient adenoviral gene delivery to mature skeletal muscle has been hindered by different factors. The low levels of adenoviral attachment receptor (CAR) that have been reported in this tissue may be a limiting factor. Therefore, adenoviral transduction of mature muscle may be improved by extending the tropism of the adenoviral vectors to attachment receptors that are highly expressed in mature myofibers. In this study, we have investigated whether an extended tropism adenoviral vector which additionally attaches to the broadly expressed heparan-containing receptors (AdPK) can bypass the maturation-dependent adenoviral transducibility of mouse skeletal muscle. METHODS The adenoviral vector AdPK carrying the LacZ gene was evaluated as a gene delivery vehicle in mouse skeletal muscle at different maturities in vitro and in vivo. The viral transduction efficiencies were determined by histochemical and ONPG analysis of the beta-galactosidase activity level. RESULTS Higher transduction efficiencies were detected in immature muscle from normal mice, and in mature muscle from merosin-deficient dy/dy mice (carrying myofibers with an impaired extracellular matrix) and dystrophin-deficient mdx mice (showing a high level of myoblast activity) when compared to mature muscle from normal mice. CONCLUSION Despite the enhanced attachment characteristics, the extended tropism adenoviral vector is, similarly to the wild-type adenoviral vector in previous studies, still hindered by both a protective extracellular matrix and the diminished myoblast-mediation in mature muscle.
Collapse
MESH Headings
- Adenoviridae/physiology
- Animals
- Cell Differentiation
- Dystrophin/deficiency
- Dystrophin/genetics
- Genes, Reporter
- Genetic Vectors/physiology
- Lac Operon
- Laminin/deficiency
- Laminin/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Mutant Strains
- Muscle Fibers, Skeletal/virology
- Muscle, Skeletal/cytology
- Muscle, Skeletal/virology
- Muscular Dystrophy, Animal/pathology
- Receptors, Virus/chemistry
- Receptors, Virus/physiology
- Recombinant Fusion Proteins/analysis
- Transfection
- beta-Galactosidase/analysis
Collapse
Affiliation(s)
- J C van Deutekom
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Children's Hospital of Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
48
|
Liu X, Brandt CR, Gabelt BT, Bryar PJ, Smith ME, Kaufman PL. Herpes simplex virus mediated gene transfer to primate ocular tissues. Exp Eye Res 1999; 69:385-95. [PMID: 10504272 DOI: 10.1006/exer.1999.0711] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We evaluated the feasibility of delivering a gene into monkey eyes using a replication-competent herpes simplex virus (HSV) type 1 ribonucleotide reductase mutant (hrR3) expressing the Escherichia coli lacZ gene. To determine the efficiency of in vitro HSV-mediated gene transfer, cultured human trabecular meshwork (HTM) and human ciliary muscle (HCM) cells were infected with hrR3 and beta-galactosidase activity was measured histochemically. Six cynomolgus monkey eyes received viral injections into the anterior chamber (2 x 10(7) pfu) and/or the vitreous (5 x 10(7) pfu), and the distribution of cells expressing lacZ was evaluated. In vitro, both cultured HTM and HCM cells displayed multiplicity-dependent beta-galactosidase activity. In vivo, intracameral and/or intravitreal injection resulted in transgene expression in TM cells and in non-pigmented ciliary epithelial cells (NPE), but not in CM cells. Transgene expression was also detected in retinal pigmented epithelial (RPE) cells and sporadic retinal ganglion cells (RGC) in eyes receiving virus intracamerally and intravitreally respectively. We observed significant inflammation in the anterior chamber, TM and CM in virus-injected eyes, along with mild vitritis and retinitis. This study demonstrates successful gene transfer using hrR3 as a vector in human ocular cells and in ocular tissues in living monkeys. Further investigation of the etiology of the inflammatory response, possible cytotoxicity, and limited duration of transgene expression is necessary in order to make this technique clinically applicable.
Collapse
Affiliation(s)
- X Liu
- Departments of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53792-3220, USA
| | | | | | | | | | | |
Collapse
|
49
|
Fechner H, Haack A, Wang H, Wang X, Eizema K, Pauschinger M, Schoemaker R, Veghel R, Houtsmuller A, Schultheiss HP, Lamers J, Poller W. Expression of coxsackie adenovirus receptor and alphav-integrin does not correlate with adenovector targeting in vivo indicating anatomical vector barriers. Gene Ther 1999; 6:1520-35. [PMID: 10490761 DOI: 10.1038/sj.gt.3301030] [Citation(s) in RCA: 243] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Recombinant adenoviral vectors are broadly applied in gene therapy protocols. However, adenovector-mediated gene transfer has limitations in vivo. One of these is the low gene transfer rate into organs other than the liver after systemic intravenous vector injection. Local direct injection into the target organ has been used as one possible solution, but increases necessary equipment and methodology and is traumatic to the target. Wild-type adenovirus infection as well as adenovector-mediated gene transfer depends on virus interaction with the Coxsackie adenovirus receptor (CAR) mediating virus attachment to the cell surface, and on interaction with alphavbeta3 and alphavbeta5 integrins mediating virus entry into the cell. In order to assess the receptor-associated potential of different tissues to act as adenovector targets, we have therefore determined CAR and alphav-integrin expression in multiple organs from different species. In addition, we have newly determined several human, rat, pig and dog CAR-mRNA sequences. Sequence comparison and structural analyses of known and of newly determined sequences suggests a potential adenovirus binding site between amino acids 29 and 128 of the CAR. With respect to the virus receptor expression patterns we found that CAR-mRNA expression was extremely variable between different tissues, with the highest levels in the liver, whereas alphav-integrin expression was far more homogenous among different organs. Both CAR and alphav-integrin showed similar expression patterns among different species. There was no correlation, however, between the adenovector expression patterns after intravenous, intracardiac and aortic root injection, respectively, and the virus receptor patterns. In summary, many organs carry both receptors required to make them potential adenovector targets. In sharp contrast, their actual targeting clearly indicates that adenovirus receptor expression is necessary but not sufficient for vector transfer after systemic injection. The apparently very important role of anatomical barriers, in particular the endothelium, requires close attention when developing non-traumatic, organ-specific gene therapy protocols.
Collapse
Affiliation(s)
- H Fechner
- Department of Cardiology and Pneumology, University Hospital Benjamin Franklin, Freie Universität, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yeung SN, Bockhold K, Tufaro F. Efficient infection of mature skeletal muscle with herpes simplex virus vectors by using dextran sulfate as a co-receptor. Gene Ther 1999; 6:1536-44. [PMID: 10490762 DOI: 10.1038/sj.gt.3300980] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The use of herpes simplex virus (HSV) vectors for gene delivery to skeletal muscle is hampered by a maturation-dependent loss of muscle fiber infectivity. Previous studies of HSV type 1 (HSV-1) infection in the rodent show that the loss of infectivity may be due, at least in part, to the development of the basal lamina throughout the course of maturation, which may block the initial events in HSV infection. To initiate infection, HSV normally attaches to cell surface heparan sulfate, which stabilizes the virus such that it can interact with secondary protein receptors required for entry into host cells. In this study, we demonstrate that heparan sulfate biosynthesis is downregulated during skeletal muscle maturation. When myofibers were treated with a variety of enzymes, including collagenase type IV or chondroitin ABC lyase, HSV infection was restored, which suggests that virus secondary receptors were present but not readily accessible to the virus in the intact myofiber. Surprisingly, we also found that HSV-1 infectivity could be restored in vitro and in vivo by exposing myofibers to low concentrations of the glycosaminoglycan analog dextran sulfate, which appears to act as a surrogate receptor to stabilize the virus at the myofiber surface such that HSV can engage additional receptors. This demonstration that the basal lamina is not an absolute block to HSV-1 infection is remarkable because it allows for the nondestructive targeting of HSV-1 to mature myofibers and greatly expands the usefulness of HSV as a gene therapy vector for the treatment of inherited and acquired diseases.
Collapse
Affiliation(s)
- S N Yeung
- Department of Microbiology and Immunology, University of British Columbia, 300-6174 University Boulevard, Vancouver, BC Canada, V6T 1Z3
| | | | | |
Collapse
|