1
|
Mern DS, Thomé C. Collagen II enrichment through scAAV6-RNAi-mediated inhibition of matrix-metalloproteinases 3 and 13 in degenerative nucleus-pulposus cells degenerative disc disease and biological treatment strategies. Exp Biol Med (Maywood) 2024; 249:10048. [PMID: 39286594 PMCID: PMC11402661 DOI: 10.3389/ebm.2024.10048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Intervertebral disc (IVD) degeneration damaging the extracellular matrix (ECM) of IVDs is the main cause of spine-associated disorders. Degenerative disc disease (DDD) is a multifaceted disorder, where environmental factors, inflammatory cytokines and catabolic enzymes act together. DDD starts typically due to imbalance between ECM biosynthesis and degradation within IVDs, especially through unbalanced degradation of aggrecan and collagen II in nucleus pulposus (NP). Current treatment approaches are primarily based on conservative or surgical therapies, which are insufficient for biological regeneration. The disintegrins and metalloproteinases with thrombospondin motifs (ADAMTSs) and matrix metalloproteinases (MMPs) are the key proteolytic enzymes for degradation of aggrecan and collagens. Previously, high expression levels of ADAMTS4, ADAMTS5, MMP3 and MMP13, which are accompanied with low levels of aggrecan and collagen II, were demonstrated in degenerative human NP cells. Moreover, self-complementary adeno-associated virus type 6 (scAAV6) mediated inhibitions of ADAMTS4 and ADAMTS5 by RNA-interference (RNAi) could specifically enhance aggrecan level. Thus, MMPs are apparently the main degrading enzymes of collagen II in NP. Furthermore, scAAV6-mediated inhibitions of MMP3 and MMP13 have not yet been investigated. Therefore, we attempted to enhance the level of collagen II in degenerative NP cells by scAAV6-RNAi-mediated inhibitions of MMP3 and MMP13. MRI was used to determine preoperative grading of IVD degeneration in patients. After isolation and culturing of NP cells, cells were transduced with scAAV6-shRNAs targeting MMP3 or MMP13; and analysed by fluorescence microscopy, FACS, MTT assay, RT-qPCR, ELISA and western blotting. scAAV6-shRNRs have no impact on cell viability and proliferation, despite high transduction efficiencies (98.6%) and transduction units (1383 TU/Cell). Combined knockdown of MMP3 (92.8%) and MMP13 (90.9%) resulted in highest enhancement of collagen II (143.2%), whereby treatment effects were significant over 56 days (p < 0.001). Conclusively, scAAV6-RNAi-mediated inhibitions of MMP3 and MMP13 help to progress less immunogenic and enduring biological treatments in DDD.
Collapse
Affiliation(s)
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Shenegelegn Mern D, Thomé C. Synergetic enrichment of aggrecan in nucleus pulposus cells by scAAV6-shRNA-mediated knockdown of aggrecanase-1 and aggrecanase-2. Exp Biol Med (Maywood) 2023; 248:1134-1144. [PMID: 37354087 PMCID: PMC10583755 DOI: 10.1177/15353702231171905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/07/2023] [Indexed: 06/26/2023] Open
Abstract
Degenerative disk disease (DDD) that aggravates structural deterioration of intervertebral disks (IVDs) can be accompanied by painful inflammation and immunopathological progressions. Current surgical or pharmacological therapies cannot repair the structure and function of IVDs. Nucleus pulposus (NP) cells are crucial for the preservation or restoration of IVDs by balancing the anabolic and catabolic factors affecting the extracellular matrix. Imbalanced anabolic and catabolic factors cause increased degradation of aggrecan. Aggrecanases A Disintegrin And Metalloproteinase with ThromboSpondin motifs (ADAMTS)4 and ADAMTS5 are the main degrading enzymes of aggrecan. Previously, we characterized adeno-associated virus (AAV6) as the most suitable serotype with marked NP cellular tropism and demonstrated that ADAMTS4 could be silenced by self-complementary adeno-associated virus grade 6 small helix ribonucleic acid (scAAV6-shRNA) in NP cells of degeneration grade III, which resulted in enrichment of aggrecan. Nonetheless, neither scAAV6-shRNA-mediated inhibition of ADAMTS5 nor joint inhibitions of ADAMTS4 and ADAMTS5 have been investigated, although both enzymes are regulated by analogous proinflammatory cytokines and have the same cleavage sites in aggrecan. Therefore, we attempted scAAV6-shRNA-mediated inhibitions of both enzymes in NP cells of degeneration grade IV to increase efficacies in treatments of DDD. The degeneration grade of IVDs in patients was determined by magnetic resonance imaging (MRI) before surgical operations. After isolation and culturing of NP cells, cells were transduced with scAAV6-shRNAs targeting ADAMTS4 or ADAMTS5. Transduced cells were analyzed by reverse transcription quantitative polymerase chain reaction (RT-qPCR), fluorescence microscopy, flow cytometry-assisted cell sorting (FACS), MTT assay (3-(4, 5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assay), immunoblotting, and enzyme-linked immunosorbent assay (ELISA). Joint transduction of NP cells exhibited high transduction efficacies (98.1%), high transduction units (TU) (1381 TU/Cell), and no effect on cell viability or proliferation. Above all joint treatments resulted in effective knockdown of ADAMTS4 (92.8%) and ADAMTS5 (93.4%) along with additive enrichment of aggrecan (113.9%). Treatment effects were significant for more than 56 days after transduction (P < 0.001). In conclusion, scAAV6-shRNA-mediated combined molecular therapy could be very valuable for more effective, durable, and less immunogenic treatment approaches in DDD.
Collapse
Affiliation(s)
| | - Claudius Thomé
- Department of Neurosurgery, Medical University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
3
|
PCR-Based Analytical Methods for Quantification and Quality Control of Recombinant Adeno-Associated Viral Vector Preparations. Pharmaceuticals (Basel) 2021; 15:ph15010023. [PMID: 35056080 PMCID: PMC8779925 DOI: 10.3390/ph15010023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Recombinant adeno-associated viral vectors (rAAV) represent a gene therapy tool of ever-increasing importance. Their utilization as a delivery vehicle for gene replacement, silencing and editing, among other purposes, demonstrate considerable versatility. Emerging vector utilization in various experimental, preclinical and clinical applications establishes the necessity of producing and characterizing a wide variety of rAAV preparations. Critically important characteristics concerning quality control are rAAV titer quantification and the detection of impurities. Differences in rAAV constructs necessitate the development of highly standardized quantification assays to make direct comparisons of different preparations in terms of assembly or purification efficiency, as well as experimental or therapeutic dosages. The development of universal methods for impurities quantification is rather complicated, since variable production platforms are utilized for rAAV assembly. However, general agreements also should be achieved to address this issue. The majority of methods for rAAV quantification and quality control are based on PCR techniques. Despite the progress made, increasing evidence concerning high variability in titration assays indicates poor standardization of the methods undertaken to date. This review summarizes successes in the field of rAAV quality control and emphasizes ongoing challenges in PCR applications for rAAV characterization. General considerations regarding possible solutions are also provided.
Collapse
|
4
|
Abstract
The adeno-associated viral vector (AAV) platform has developed into a primary modality for efficient in vivo, and in more limited settings, in vitro or ex vivo gene transfer. Its applications range from a tool for experimental purposes to preclinical and clinical gene therapy. The ability to accurately and reproducibly quantify vector concentration is critical for any of these applications. While several quantification assays are available, here we outline a detailed protocol for the quantification of DNase-I protected vector genomes reliant on the polymerase chain reaction (PCR) as a measure of the active component of the vector, namely its transgene cargo. With the emergence of droplet digital PCR (ddPCR), we provide side-by-side protocols for traditional TaqMan™ real-time, quantitative PCR (qPCR) and ddPCR, as well as comparative data generated with both methods. Lastly, we discuss the importance of the use of surfactant (here, Pluronic® F-68) in the execution of the assay to limit DNA and AAV adherence to various carriers during the titration, particularly at low concentrations. We believe these protocols can lead to reduced variability and increased comparability between AAV studies.
Collapse
|
5
|
Hiller T, Berg J, Elomaa L, Röhrs V, Ullah I, Schaar K, Dietrich AC, Al-Zeer MA, Kurtz A, Hocke AC, Hippenstiel S, Fechner H, Weinhart M, Kurreck J. Generation of a 3D Liver Model Comprising Human Extracellular Matrix in an Alginate/Gelatin-Based Bioink by Extrusion Bioprinting for Infection and Transduction Studies. Int J Mol Sci 2018; 19:E3129. [PMID: 30321994 PMCID: PMC6213460 DOI: 10.3390/ijms19103129] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/27/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023] Open
Abstract
Bioprinting is a novel technology that may help to overcome limitations associated with two-dimensional (2D) cell cultures and animal experiments, as it allows the production of three-dimensional (3D) tissue models composed of human cells. The present study describes the optimization of a bioink composed of alginate, gelatin and human extracellular matrix (hECM) to print human HepaRG liver cells with a pneumatic extrusion printer. The resulting tissue model was tested for its suitability for the study of transduction by an adeno-associated virus (AAV) vector and infection with human adenovirus 5 (hAdV5). We found supplementation of the basic alginate/gelatin bioink with 0.5 and 1 mg/mL hECM provides desirable properties for the printing process, the stability of the printed constructs, and the viability and metabolic functions of the printed HepaRG cells. The tissue models were efficiently transduced by AAV vectors of serotype 6, which successfully silenced an endogenous target (cyclophilin B) by means of RNA interference. Furthermore, the printed 3D model supported efficient adenoviral replication making it suitable to study virus biology and develop new antiviral compounds. We consider the approach described here paradigmatic for the development of 3D tissue models for studies including viral vectors and infectious viruses.
Collapse
Affiliation(s)
- Thomas Hiller
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Johanna Berg
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Department of Organic Chemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| | - Viola Röhrs
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Imran Ullah
- Berlin-Brandenburger Centrum für Regenerative Therapien, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Katrin Schaar
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Ann-Christin Dietrich
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Munir A Al-Zeer
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Andreas Kurtz
- Berlin-Brandenburger Centrum für Regenerative Therapien, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Andreas C Hocke
- Dept. of Internal Medicine/Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Stefan Hippenstiel
- Dept. of Internal Medicine/Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Henry Fechner
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Department of Organic Chemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| | - Jens Kurreck
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany.
| |
Collapse
|
6
|
Valdor M, Wagner A, Röhrs V, Berg J, Fechner H, Schröder W, Tzschentke TM, Bahrenberg G, Christoph T, Kurreck J. RNA interference-based functional knockdown of the voltage-gated potassium channel Kv7.2 in dorsal root ganglion neurons after in vitro and in vivo gene transfer by adeno-associated virus vectors. Mol Pain 2017; 14:1744806917749669. [PMID: 29212407 PMCID: PMC5805000 DOI: 10.1177/1744806917749669] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activation of the neuronal potassium channel Kv7.2 encoded by the KCNQ2 gene has recently been shown to be an attractive mechanism to inhibit nociceptive transmission. However, potent, selective, and clinically proven activators of Kv7.2/Kv7.3 currents with analgesic properties are still lacking. An important prerequisite for the development of new drugs is a model to test the selectivity of novel agonists by abrogating Kv7.2/Kv7.3 function. Since constitutive knockout mice are not viable, we developed a model based on RNA interference-mediated silencing of KCNQ2. By delivery of a KCNQ2-specific short hairpin RNA with adeno-associated virus vectors, we completely abolished the activity of the specific Kv7.2/Kv7.3-opener ICA-27243 in rat sensory neurons. Results obtained in the silencing experiments were consistent between freshly prepared and cryopreserved dorsal root ganglion neurons, as well as in dorsal root ganglion neurons dissociated and cultured after in vivo administration of the silencing vector by intrathecal injections into rats. Interestingly, the tested associated virus serotypes substantially differed with respect to their transduction capability in cultured neuronal cell lines and primary dorsal root ganglion neurons and the in vivo transfer of transgenes by intrathecal injection of associated virus vectors. However, our study provides the proof-of-concept that RNA interference-mediated silencing of KCNQ2 is a suitable approach to create an ex vivo model for testing the specificity of novel Kv7.2/Kv7.3 agonists.
Collapse
Affiliation(s)
- Markus Valdor
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Anke Wagner
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Viola Röhrs
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Johanna Berg
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Henry Fechner
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Wolfgang Schröder
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Thomas M Tzschentke
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | | | - Thomas Christoph
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Jens Kurreck
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| |
Collapse
|
7
|
Liu D, Mei X, Wang L, Yang X. RhoA inhibits apoptosis and increases proliferation of cultured SPCA1 lung cancer cells. Mol Med Rep 2017; 15:3963-3968. [PMID: 28487954 PMCID: PMC5436147 DOI: 10.3892/mmr.2017.6545] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 01/18/2017] [Indexed: 01/08/2023] Open
Abstract
The Rho kinase pathway has previously been reported to possess a close relationship with the growth, migration and invasion of lung cancer cells. However, the molecular mechanisms underlying the effects of this pathway on lung cancer cells are still elusive. The aim of the present study was to investigate the effects and underlying molecular mechanisms of Ras homolog family member A (RhoA) on the proliferation and apoptosis of SPCA1 lung carcinoma cells. Stable SPCA1 lung cancer cell lines, in which RhoA expression was silenced by small interfering RNA, were isolated following Geneticin screening. Inhibition of RhoA expression significantly decreased the proliferation of SPCA1 lung cancer cells, whereas apoptosis was significantly increased (P<0.01) as determined by the MTS tetrazolium assay and flow cytometry analysis, respectively. At the molecular level, knockdown of RhoA resulted in the significant activation of caspase-3 (P<0.01), and a significant reduction in the levels of phosphorylated signal transducer and activator of transcription (phospho-STAT3; P<0.01), as determined by western blotting. The results suggested that RhoA knockdown prevents cell proliferation and induces apoptosis in SPCA1 lung cancer cells. Furthermore, the underlying mechanisms responsible for these effects may include the activation of caspase-3 and the reduction of phospho-STAT3 levels.
Collapse
Affiliation(s)
- Dabei Liu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Xingke Mei
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Linlin Wang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Xueying Yang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
8
|
Shenegelegn Mern D, Tschugg A, Hartmann S, Thomé C. Self-complementary adeno-associated virus serotype 6 mediated knockdown of ADAMTS4 induces long-term and effective enhancement of aggrecan in degenerative human nucleus pulposus cells: A new therapeutic approach for intervertebral disc disorders. PLoS One 2017; 12:e0172181. [PMID: 28207788 PMCID: PMC5313142 DOI: 10.1371/journal.pone.0172181] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/31/2017] [Indexed: 01/07/2023] Open
Abstract
Inhibition of intervertebral disc (IVD) degeneration, which is often accompanied by painful inflammatory and immunopathological processes, is challenging. Current IVD gene therapeutic approaches are based on adenoviral gene delivery systems, which are limited by immune reactions to their viral proteins. Their applications in IVDs near to sensitive neural structure could provoke toxicity and immunological side-effects with neurological deficits. Self-complementary adeno-associated virus (scAAV) vectors, which do not express any viral gene and are not linked with any known disease in humans, are attractive therapeutic gene delivery vectors in degenerative IVDs. However, scAAV-based silencing of catabolic or inflammatory factor has not yet been investigated in human IVD cells. Therefore, we used scAAV6, the most suitable serotype for transduction of human nucleus pulposus (NP) cells, to knockdown the major catabolic gene (ADAMTS4) of IVD degeneration. IVD degeneration grades were determined by preoperative magnetic resonance imaging. Lumbar NP tissues of degeneration grade III were removed from 12 patients by nucleotomy. NP cells were isolated and cultured with low-glucose. Titre of recombinant scAAV6 vectors targeting ADAMTS4, transduction efficiencies, transduction units, cell viabilities and expression levels of target genes were analysed using quantitative PCR, fluorescence microscopy, fluorescence-activated cell sorting, 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assays, quantitative reverse transcription PCR, western blot and enzyme-linked immunosorbent assays during 48 days of post-transduction. Transduction efficiencies between 98.2% and 37.4% and transduction units between 611 and 245 TU/cell were verified during 48 days of post-transduction (p<0.001). scAAV6-mediated knockdown of ADAMTS4 with maximum 87.7% and minimum 40.1% was confirmed on day 8 and 48 with enhanced the level of aggrecan 48.5% and 30.2% respectively (p<0.001). scAAV6-mediated knockdown of ADAMTS4 showed no impact on cell viability and expression levels of other inflammatory catabolic proteins. Thus, our results are promising and may help to design long-term and less immunogenic gene therapeutic approaches in IVD disorders, which usually need prolonged therapeutic period between weeks and months.
Collapse
Affiliation(s)
| | - Anja Tschugg
- Department of Neurosurgery, Innsbruck Medical University, Innsbruck, Austria
| | - Sebastian Hartmann
- Department of Neurosurgery, Innsbruck Medical University, Innsbruck, Austria
| | - Claudius Thomé
- Department of Neurosurgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
9
|
Hiller T, Röhrs V, Dehne EM, Wagner A, Fechner H, Lauster R, Kurreck J. Study of Viral Vectors in a Three-dimensional Liver Model Repopulated with the Human Hepatocellular Carcinoma Cell Line HepG2. J Vis Exp 2016. [PMID: 27805597 PMCID: PMC5092236 DOI: 10.3791/54633] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This protocol describes the generation of a three-dimensional (3D) ex vivo liver model and its application to the study and development of viral vector systems. The model is obtained by repopulating the extracellular matrix of a decellularized rat liver with a human hepatocyte cell line. The model permits studies in a vascularized 3D cell system, replacing potentially harmful experiments with living animals. Another advantage is the humanized nature of the model, which is closer to human physiology than animal models. In this study, we demonstrate the transduction of this liver model with a viral vector derived from adeno-associated viruses (AAV vector). The perfusion circuit that supplies the 3D liver model with media provides an easy means to apply the vector. The system permits monitoring of the major metabolic parameters of the liver. For final analysis, tissue samples can be taken to determine the extent of recellularization by histological techniques. Distribution of the virus vector and expression of the delivered transgene can be analyzed by quantitative PCR (qPCR), Western blotting and immunohistochemistry. Numerous applications of the vector model in basic research and in the development of gene therapeutic applications can be envisioned, including the development of novel antiviral therapeutics, cancer research, and the study of viral vectors and their potential side effects.
Collapse
Affiliation(s)
- Thomas Hiller
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology
| | - Viola Röhrs
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology
| | - Eva-Maria Dehne
- Department of Medical Biotechnology, Institute of Biotechnology, Berlin University of Technology
| | - Anke Wagner
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology; Department of Bioprocess Engineering, Institute of Biotechnology, Berlin University of Technology
| | - Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology
| | - Roland Lauster
- Department of Medical Biotechnology, Institute of Biotechnology, Berlin University of Technology
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology;
| |
Collapse
|
10
|
Mern DS, Thomé C. Identification and characterization of human nucleus pulposus cell specific serotypes of adeno-associated virus for gene therapeutic approaches of intervertebral disc disorders. BMC Musculoskelet Disord 2015; 16:341. [PMID: 26552484 PMCID: PMC4640218 DOI: 10.1186/s12891-015-0799-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022] Open
Abstract
Background Intervertebral disc (IVD) disorders are often accompanied by painful inflammatory and immunopathological processes. Nucleus pulposus (NP) cells play a pivotal role in maintenance of IVD by organizing the expression of anabolic, catabolic, anti-catabolic and inflammatory cytokines. Human NP cells have been targeted by gene therapeutic approaches using lentiviral or adenoviral systems that could be critical due to genome incorporation or immunological side effects. Adeno-associated viruses (AAVs), which do not express any viral gene and are not linked with any known disease in humans, are attractive gene delivery vectors. However, their lack of specific tissue tropism and preexisting immune response are main problems for therapeutic applications. Heretofore, AAVs have not been studied in human IVD research. Therefore, we attempted to identify NP cell specific AAV serotype by targeting human NP cells with different self-complementary AAV (scAAV) serotypes. Identification and characterization of the proper serotype is crucial to establish less immunogenic and safer gene therapeutic approaches of IVD disorders. Methods Preoperative magnetic resonance imaging (MRI) was used for grading of IVD degeneration. NP cells were isolated, cultured with low-glucose and transduced with green fluorescent protein (GFP) packing scAAV serotypes (scAAV1-8) in a dose-dependent manner. scAAV titers were determined by quantitative polymerase chain reaction (qPCR). Transduction efficiencies were determined by fluorescence microscopy and fluorescence-activated cell sorting within 48 days of post-transduction. The 3-(4, 5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay was used to determine NP cell viability. Three-dimensional (3D) cell culture and enzyme-linked immunosorbant assay (ELISA) were performed to examine the expression levels of inflammatory, catabolic and matrix proteins in NP cells. Results scAAV6, scAAV2 and scAAV3 showed high and prolonged transgene GFP expressions with transdution efficiencies of 98.6 %, 91.5 % and 89.6 % respectively (p ≤ 0.002). Unlike scAAV6, the serotypes scAAV2 and scAAV3 declined the viability of NP cells by about 25 % and 10 % respectively (p ≤ 0.001). Moreover, scAAV6 did not affect the expression of the inflammatory, catabolic and matrix proteins. Conclusions As original primary research evaluating AAVs in degenerative human IVDs, this study identified scAAV6 as a proper serotype for high, stable and non-immunogenic target gene expression in human NP cells. The data could be very important to design efficient and safer gene therapeutic approaches of IVD disorders.
Collapse
Affiliation(s)
- Demissew S Mern
- Department of Neurosurgery, Innsbruck Medical University, Anichstrasse 35, Innsbruck, A-6020, Austria.
| | - Claudius Thomé
- Department of Neurosurgery, Innsbruck Medical University, Anichstrasse 35, Innsbruck, A-6020, Austria.
| |
Collapse
|
11
|
Piedra J, Ontiveros M, Miravet S, Penalva C, Monfar M, Chillon M. Development of a rapid, robust, and universal picogreen-based method to titer adeno-associated vectors. Hum Gene Ther Methods 2015; 26:35-42. [PMID: 25640021 DOI: 10.1089/hgtb.2014.120] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) are promising vectors in preclinical and clinical assays for the treatment of diseases with gene therapy strategies. Recent technological advances in amplification and purification have allowed the production of highly purified rAAV vector preparations. Although quantitative polymerase chain reaction (qPCR) is the current method of choice for titrating rAAV genomes, it shows high variability. In this work, we report a rapid and robust rAAV titration method based on the quantitation of encapsidated DNA with the fluorescent dye PicoGreen®. This method allows detection from 3×10(10) viral genome/ml up to 2.4×10(13) viral genome/ml in a linear range. Contrasted with dot blot or qPCR, the PicoGreen-based assay has less intra- and interassay variability. Moreover, quantitation is rapid, does not require specific primers or probes, and is independent of the rAAV pseudotype analyzed. In summary, development of this universal rAAV-titering method may have substantive implications in rAAV technology.
Collapse
Affiliation(s)
- Jose Piedra
- 1 Vector Production Unit, Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona , Bellaterra 08193, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
12
|
Wagner A, Röhrs V, Materne EM, Hiller T, Kedzierski R, Fechner H, Lauster R, Kurreck J. Use of a three-dimensional humanized liver model for the study of viral gene vectors. J Biotechnol 2015; 212:134-43. [PMID: 26356676 DOI: 10.1016/j.jbiotec.2015.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 07/17/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
Abstract
Reconstituted three-dimensional (3D) liver models obtained by engrafting hepatic cells into an extracellular matrix (ECM) are valuable tools to study tissue regeneration, drug action and toxicology ex vivo. The aim of the present study was to establish a system for the functional investigation of a viral vector in a 3D liver model composed of human HepG2 cells on a rat ECM. An adeno-associated viral (AAV) vector expressing the Emerald green fluorescent protein (EmGFP) and a short hairpin RNA (shRNA) directed against human cyclophilin b (hCycB) was injected into the portal vein of 3D liver models. Application of the vector did not exert toxic effects, as shown by analysis of metabolic parameters. Six days after transduction, fluorescence microscopy analysis of EmGFP production revealed widespread distribution of the AAV vectors. After optimization of the recellularization and transduction conditions, averages of 55 and 90 internalized vector genomes per cell in two replicates of the liver model were achieved, as determined by quantitative PCR analysis. Functionality of the AAV vector was confirmed by efficient shRNA-mediated knockdown of hCycB by 70-90%. Our study provides a proof-of-concept that a recellularized biological ECM provides a valuable model to study viral vectors ex vivo.
Collapse
Affiliation(s)
- Anke Wagner
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Viola Röhrs
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Eva-Maria Materne
- Department of Medical Biotechnology, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Thomas Hiller
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Radoslaw Kedzierski
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Henry Fechner
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Roland Lauster
- Department of Medical Biotechnology, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany.
| |
Collapse
|