1
|
Islam T, Oh J, Fortin L, Connors C, Deorkar N. Streamlining viral safety in AAV manufacturing: simultaneous cell lysis and viral inactivation using biodegradable detergents. J Pharm Sci 2025; 114:103761. [PMID: 40107420 DOI: 10.1016/j.xphs.2025.103761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Ensuring viral safety in biopharmaceutical production is vital for global regulatory standards adherence and safeguarding patients. This study evaluates the efficacy of newly developed biodegradable detergent formulations, designed to comply with regulatory requirements, for inactivating enveloped viruses during the cell lysis step of adeno-associated virus (AAV) production. Virus clearance (VC) studies showed robust inactivation kinetics against Xenotropic Murine Leukemia Virus (XMuLV), achieving a log reduction value (LRV) of ≥5.34 within 30 to 120 min. Comparative testing in crude cell lysate and PBS buffer showed consistent viral inactivation across different matrices, highlighting the versatility of the detergent formulations. Importantly, the detergent composition preserved AAV capsid integrity, and subsequent purification steps, including affinity and ion-exchange chromatography, effectively removed residual detergent. Overall, this study demonstrates the potential of biodegradable detergents to simultaneously facilitate cell lysis and viral inactivation in AAV manufacturing, offering a sustainable alternative that aligns with industry standards.
Collapse
Affiliation(s)
- Tuhidul Islam
- Biopharma Production, Avantor Inc., Bridgewater, NJ, United States
| | - Jungmin Oh
- Biopharma Production, Avantor Inc., Bridgewater, NJ, United States
| | - Lori Fortin
- Biopharma Production, Avantor Inc., Bridgewater, NJ, United States
| | - Courtney Connors
- Biopharma Production, Avantor Inc., Bridgewater, NJ, United States
| | | |
Collapse
|
2
|
Singh S, Khan S, Khan S, Ansari O, Malhotra N, Shukla SK, Narang J. Muscle Matters: Transforming Amyotrophic Lateral Sclerosis Diagnostics with Next-Gen Biosensors and Smart Detection. ACS Chem Neurosci 2025; 16:563-587. [PMID: 39910731 DOI: 10.1021/acschemneuro.4c00664] [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] [Indexed: 02/07/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that primarily targets the motor system, causing patients' speech and swallowing ability to rapidly deteriorate. Although ALS is usually classified into familial and sporadic forms, diagnosing it can be extremely difficult due to the absence of definitive biomarkers, often resulting in delays in diagnosis. Current diagnostic practices rely heavily on clinical assessments that indicate damage to both upper motor neurons (UMNs) and lower motor neurons (LMNs). This includes comprehensive physical examinations, electromyography (EMG) to assess neuromuscular function, and the exclusion of other similar conditions such as cervical spondylotic myelopathy, multifocal motor neuropathy, and Kennedy's disease through appropriate diagnostic procedures. The urgent need for specific biomarkers is critical for timely diagnosis and therapeutic advancements in ALS management. While many recent developments in research have not yet translated into direct patient benefits, the recognition of ALS as a complex disease is beginning to influence clinical practice significantly. Optimal management strategies emphasize on symptom control and improving the quality of life for patients within a holistic healthcare framework. This review provides a comprehensive overview of ALS, delving into its pathophysiology, clinical symptoms, and the latest advancements in detection methods that utilize traditional approaches, innovative biosensors, and smart diagnostic technologies. It discusses various treatment options available for ALS while exploring future developments that may enhance patient screening and improve clinical outcomes. By integrating assessments into the underlying mechanisms of the disease with cutting-edge diagnostic approaches, this review aims to contribute meaningfully to ongoing efforts to optimize ALS management and therapeutic strategies, ultimately improving patient care and outcomes.
Collapse
Affiliation(s)
- Saumitra Singh
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India
| | - Sameer Khan
- Department of Biochemistry, Jamia Hamdard, New Delhi 110062, India
| | - Shina Khan
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India
| | - Osheen Ansari
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India
| | - Nitesh Malhotra
- Department of Physiotherapy, School of Allied Health Science, Manav Rachna International Institute of Research and Studies, Faridabad 121004, India
| | - Sudheesh K Shukla
- Centre for Nanoscience and Nano bioelectronics, School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Jagriti Narang
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
3
|
Akinyele O, Tran KB, Johnson MA, Kemaladewi DU. Gene replacement therapy to restore polyamine metabolism in a Snyder-Robinson syndrome mouse model. Methods Enzymol 2025; 715:271-292. [PMID: 40382143 DOI: 10.1016/bs.mie.2025.01.068] [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] [Indexed: 05/20/2025]
Abstract
Polyamines, including putrescine, spermidine, and spermine, are organic cations essential for cell growth, proliferation, and tissue regeneration. Their levels are tightly regulated by a set of enzymes controlling their biosynthesis, catabolism, and interconversion. Dysregulation of polyamine metabolism is associated with a group of rare genetic neurodevelopmental disorders collectively known as "polyaminopathies", including Snyder-Robinson Syndrome (SRS). SRS is an X-linked recessive disorder caused by mutations in the SMS gene, which encodes the spermine synthase enzyme. The lack of spermine synthase leads to aberrant polyamine levels and neurological impairments, as observed in patients and animal models. Currently, there are no available treatment options for SRS. Due to its monogenic nature, SRS is an excellent candidate for gene replacement therapy. The recent success of Zolgensma in treating children with Spinal Muscular Atrophy and the establishment of Platform Vector Gene Therapy (Pave-GT) initiative at the National Institute of Health (NIH) offer a framework to adapt-and-apply the same gene delivery system for multiple rare disease gene therapies. This chapter outlines strategies for delivering a functional copy of the SMS gene using an adeno-associated viral (AAV) vector, as well as methods to evaluate the molecular efficacy of this approach in an SRS mouse model. Our ultimate goal is to establish a versatile platform for genetic interventions targeting SRS and other polyaminopathies.
Collapse
Affiliation(s)
- Oluwaseun Akinyele
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Krystal B Tran
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Marie A Johnson
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Dwi U Kemaladewi
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
4
|
Burke M, Blatt B, Teixeira J, Pérez‐López D, Yue Y, Pan X, Hakim C, Yao G, Herzog R, Duan D. Adeno-Associated Virus 8 and 9 Myofibre Type/Size Tropism Profiling Reveals Therapeutic Effect of Microdystrophin in Canines. J Cachexia Sarcopenia Muscle 2025; 16:e13681. [PMID: 39790021 PMCID: PMC11718217 DOI: 10.1002/jcsm.13681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/08/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) 8 and 9 are in clinical trials for treating neuromuscular diseases such as Duchenne muscular dystrophy (DMD). Muscle consists of myofibres of different types and sizes. However, little is known about the fibre type and fibre size tropism of AAV in large mammals. METHODS We evaluated fibre type- and size-specific transduction properties of AAV8 and AAV9 in 17 dogs that received systemic gene transfer (dose 1.94 ± 0.52 × 1014 vg/kg; injected at 2.86 ± 0.30 months; harvested at 20.79 ± 3.30 months). For AAV8, two DMD dogs and three carrier dogs received an alkaline phosphatase (AP) reporter vector, and five DMD dogs received a four-repeat microdystrophin (uDys) vector. For AAV9, one normal and one DMD dog received the AP vector, and five DMD dogs received a five-repeat uDys vector. Association between AAV transduction and the fibre type/size was studied in three muscles that showed mosaic transgene expression, including the biceps femoris, teres major and latissimus dorsi. RESULTS Transgene expression was detected in 30%-45% of myofibres. In the AP reporter vector-injected dogs, neither AAV8 nor AAV9 showed a statistically significant fibre type preference. Interestingly, AP expression was enriched in smaller fibres. In uDys-treated DMD dogs, slow and fast myofibres were equally transduced. Notably, uDys-expressing myofibres were significantly larger than uDys-negative myofibres irrespective of the AAV serotype (p < 0.0001). In AAV8 uDys vector-injected dogs, the mini-Feret diameter was 15%, 16% and 23% larger in uDys-positive slow, fast and hybrid fibres, respectively; the cross-sectional area was 30%, 34% and 46% larger in uDys-positive slow, fast and hybrid fibres, respectively. In AAV9 uDys vector-injected dogs, the mini-Feret diameter was 12%, 13% and 25% larger in uDys-positive slow, fast and hybrid fibres, respectively; the cross-sectional area was 25%, 28% and 59% larger in uDys-positive slow, fast and hybrid fibres, respectively. CONCLUSIONS Our studies suggest that AAV8 and AAV9 transduce fast and slow myofibres at equivalent efficiency. Importantly, uDys therapy effectively prevented dystrophic myofibre atrophy. Our study provides important insight into systemic muscle AAV delivery in large mammals and supports further development of uDys gene therapy for DMD.
Collapse
Affiliation(s)
- Matthew J. Burke
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Braiden M. Blatt
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
- College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - James A. Teixeira
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Dennis O. Pérez‐López
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Gang Yao
- Department of Chemical and Biomedical Engineering, College of EngineeringUniversity of MissouriColumbiaMissouriUSA
| | - Roland W. Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric ResearchIndiana UniversityIndianapolisIndianaUSA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
- Department of Chemical and Biomedical Engineering, College of EngineeringUniversity of MissouriColumbiaMissouriUSA
- Department of Biomedical Sciences, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
- Department of Neurology, School of MedicineUniversity of MissouriColumbiaMissouriUSA
| |
Collapse
|
5
|
Koh WC, Yusoff K, Song AAL, Saad N, Chia SL. Viral vectors: design and delivery for small RNA. J Med Microbiol 2025; 74. [PMID: 39950625 DOI: 10.1099/jmm.0.001972] [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] [Indexed: 02/18/2025] Open
Abstract
RNA interference regulates gene expression by selectively silencing target genes through the introduction of small RNA molecules, such as microRNA, small interfering RNA and short hairpin RNA. These molecules offer significant therapeutic potential for diverse human ailments like cancer, viral infections and neurodegenerative disorders. Whilst non-viral vectors like nanoparticles have been extensively explored for delivering these RNAs, viral vectors, with superior specificity and delivery efficiency, remain less studied. This review examines current viral vectors for small RNA delivery, focusing on design strategies and characteristics. It compares the advantages and drawbacks of each vector, aiding readers in selecting the optimal one for small RNA delivery.
Collapse
Affiliation(s)
- Wei Chin Koh
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Khatijah Yusoff
- Department of Microbiology, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Malaysia Genome & Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000 Kajang, Selangor, Malaysia
| | - Adelene Ai Lian Song
- Department of Microbiology, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suet Lin Chia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology & Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Malaysia Genome & Vaccine Institute, National Institutes of Biotechnology Malaysia, Jalan Bangi, 43000 Kajang, Selangor, Malaysia
| |
Collapse
|
6
|
Kodippili K, Hakim CH, Burke MJ, Yue Y, Teixeira JA, Zhang K, Yao G, Babu GJ, Herzog RW, Duan D. SERCA2a overexpression improves muscle function in a canine Duchenne muscular dystrophy model. Mol Ther Methods Clin Dev 2024; 32:101268. [PMID: 38911286 PMCID: PMC11190715 DOI: 10.1016/j.omtm.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 06/25/2024]
Abstract
Excessive cytosolic calcium accumulation contributes to muscle degeneration in Duchenne muscular dystrophy (DMD). Sarco/endoplasmic reticulum calcium ATPase (SERCA) is a sarcoplasmic reticulum (SR) calcium pump that actively transports calcium from the cytosol into the SR. We previously showed that adeno-associated virus (AAV)-mediated SERCA2a therapy reduced cytosolic calcium overload and improved muscle and heart function in the murine DMD model. Here, we tested whether AAV SERCA2a therapy could ameliorate muscle disease in the canine DMD model. 7.83 × 1013 vector genome particles of the AAV vector were injected into the extensor carpi ulnaris (ECU) muscles of four juvenile affected dogs. Contralateral ECU muscles received excipient. Three months later, we observed widespread transgene expression and significantly increased SERCA2a levels in the AAV-injected muscles. Treatment improved SR calcium uptake, significantly reduced calpain activity, significantly improved contractile kinetics, and significantly enhanced resistance to eccentric contraction-induced force loss. Nonetheless, muscle histology was not improved. To evaluate the safety of AAV SERCA2a therapy, we delivered the vector to the ECU muscle of adult normal dogs. We achieved strong transgene expression without altering muscle histology and function. Our results suggest that AAV SERCA2a therapy has the potential to improve muscle performance in a dystrophic large mammal.
Collapse
Affiliation(s)
- Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Matthew J. Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - James A. Teixeira
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Gang Yao
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
7
|
Wang H, Kwak SE, Zheng A, Arias EB, Pan X, Duan D, Cartee GD. Phosphorylation of AS160-serine 704 is not essential for exercise-increase in insulin-stimulated glucose uptake by skeletal muscles from female or male rats. Am J Physiol Endocrinol Metab 2024; 326:E807-E818. [PMID: 38656130 PMCID: PMC11376492 DOI: 10.1152/ajpendo.00010.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle from rodents and humans of both sexes. We recently found that concurrent mutation of three key sites to prevent their phosphorylation (Ser588, Thr642, and Ser704) on Akt substrate of 160 kDa (AS160; also known as TBC1D4) reduced the magnitude of the enhancement of postexercise ISGU (PEX-ISGU) by muscle from male, but not female rats. However, we did not test the role of individual phosphorylation sites on PEX-ISGU. Accordingly, our current aim was to test whether AS160 Ser704 phosphorylation (pSer704) is required for elevated PEX-ISGU by muscle. AS160-knockout (AS160-KO) rats (female and male) were studied when either in sedentary or 3 h after acute exercise. Adeno-associated virus (AAV) vectors were used to enable muscle expression of wild-type AS160 (AAV-WT-AS160) or AS160 mutated Ser704 to alanine to prevent phosphorylation (AAV-1P-AS160). Paired epitrochlearis muscles from each rat were injected with AAV-WT-AS160 or AAV-1P-AS160. We discovered that regardless of sex 1) AS160 abundance in AS160-KO rats was similar in paired muscles expressing WT-AS160 versus 1P-AS160; 2) muscles from exercised versus sedentary rats had greater ISGU, and PEX-ISGU was slightly greater for muscles expressing 1P-AS160 versus contralateral muscles expressing WT-AS160; and 3) pAS160Thr642 was lower in muscles expressing 1P-AS160 versus paired muscles expressing WT-AS160. These results indicate that pAS160Ser704 was not essential for elevated PEX-ISGU by skeletal muscle from rats of either sex. Furthermore, elimination of the postexercise increase in pAS160Thr642 did not lessen the postexercise effect on ISGU.NEW & NOTEWORTHY The current study evaluated the role of Akt substrate of 160 kDa (AS160) phosphorylation on Ser704 in increased insulin-stimulated glucose uptake by skeletal muscle after exercise. Adeno-associated virus vectors were engineered to express either wild-type-AS160 or AS160 mutated so that it could not be phosphorylated on Ser704 in paired muscles from AS160-knockout rats. The results demonstrated that AS160 phosphorylation on Ser704 was not essential for exercise-induced elevation in insulin-stimulated glucose uptake by rats of either sex.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Chemical and Biomedical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, United States
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
8
|
Wang H, Zheng A, Arias EB, Kwak SE, Pan X, Duan D, Cartee GD. AS160 expression, but not AS160 Serine-588, Threonine-642, and Serine-704 phosphorylation, is essential for elevated insulin-stimulated glucose uptake by skeletal muscle from female rats after acute exercise. FASEB J 2023; 37:e23021. [PMID: 37289137 DOI: 10.1096/fj.202300282rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle in both sexes. We recently found that muscle expression and phosphorylation of key sites of Akt substrate of 160 kDa (AS160; also called TBC1D4) are essential for the full-exercise effect on postexercise-ISGU (PEX-ISGU) in male rats. In striking contrast, AS160's role in increased PEX-ISGU has not been rigorously tested in females. Our rationale was to address this major knowledge gap. Wild-type (WT) and AS160-knockout (KO) rats were either sedentary or acutely exercised. Adeno-associated virus (AAV) vectors were engineered to express either WT-AS160 or AS160 mutated on key serine and threonine residues (Ser588, Thr642, and Ser704) to alanine to prevent their phosphorylation. AAV vectors were delivered to the muscle of AS160-KO rats to determine if WT-AS160 or phosphorylation-inactivated AS160 would influence PEX-ISGU. AS160-KO rats have lower skeletal muscle abundance of the GLUT4 glucose transporter protein. This GLUT4 deficit was rescued using AAV delivery of GLUT4 to determine if eliminating muscle GLUT4 deficiency would normalize PEX-ISGU. The novel results were as follows: (1) AS160 expression was required for greater PEX-ISGU; (2) rescuing muscle AS160 expression in AS160-KO rats restored elevated PEX-ISGU; (3) AS160's essential role for the postexercise increase in ISGU was not attributable to reduced muscle GLUT4 content; and (4) AS160 phosphorylation on Ser588, Thr642, and Ser704 was not essential for greater PEX-ISGU. In conclusion, these novel findings revealed that three phosphosites widely proposed to influence PEX-ISGU are not required for this important outcome in female rats.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Morales ED, Yue Y, Watkins TB, Han J, Pan X, Gibson AM, Hu B, Brito‐Estrada O, Yao G, Makarewich CA, Babu GJ, Duan D. Dwarf Open Reading Frame (DWORF) Gene Therapy Ameliorated Duchenne Muscular Dystrophy Cardiomyopathy in Aged mdx Mice. J Am Heart Assoc 2023; 12:e027480. [PMID: 36695318 PMCID: PMC9973626 DOI: 10.1161/jaha.122.027480] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/21/2022] [Indexed: 01/26/2023]
Abstract
Background Cardiomyopathy is a leading health threat in Duchenne muscular dystrophy (DMD). Cytosolic calcium upregulation is implicated in DMD cardiomyopathy. Calcium is primarily removed from the cytosol by the sarcoendoplasmic reticulum calcium ATPase (SERCA). SERCA activity is reduced in DMD. Improving SERCA function may treat DMD cardiomyopathy. Dwarf open reading frame (DWORF) is a recently discovered positive regulator for SERCA, hence, a potential therapeutic target. Methods and Results To study DWORF's involvement in DMD cardiomyopathy, we quantified DWORF expression in the heart of wild-type mice and the mdx model of DMD. To test DWORF gene therapy, we engineered and characterized an adeno-associated virus serotype 9-DWORF vector. To determine if this vector can mitigate DMD cardiomyopathy, we delivered it to 6-week-old mdx mice (6×1012 vector genome particles/mouse) via the tail vein. Exercise capacity, heart histology, and cardiac function were examined at 18 months of age. We found DWORF expression was significantly reduced at the transcript and protein levels in mdx mice. Adeno-associated virus serotype 9-DWORF vector significantly enhanced SERCA activity. Systemic adeno-associated virus serotype 9-DWORF therapy reduced myocardial fibrosis and improved treadmill running, electrocardiography, and heart hemodynamics. Conclusions Our data suggest that DWORF deficiency contributes to SERCA dysfunction in mdx mice and that DWORF gene therapy holds promise to treat DMD cardiomyopathy.
Collapse
Affiliation(s)
- Emily D. Morales
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Thais B. Watkins
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Jin Han
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Aaron M. Gibson
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
| | - Bryan Hu
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
| | - Omar Brito‐Estrada
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
| | - Gang Yao
- Department of Biomedical, Biological & Chemical Engineering, College of EngineeringThe University of MissouriColumbiaMO
| | - Catherine A. Makarewich
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical CenterThe Heart InstituteCincinnatiOH
- Department of PediatricsThe University of Cincinnati College of MedicineCincinnatiOH
| | - Gopal J. Babu
- Department of Cell Biology and Molecular MedicineRutgers, New Jersey Medical SchoolNewarkNJ
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of MedicineThe University of MissouriColumbiaMO
- Department of Biomedical, Biological & Chemical Engineering, College of EngineeringThe University of MissouriColumbiaMO
- Department of Neurology, School of MedicineThe University of MissouriColumbiaMO
- Department of Biomedical Sciences, College of Veterinary MedicineThe University of MissouriColumbiaMO
| |
Collapse
|
10
|
Directed evolution of adeno-associated virus for efficient gene delivery to microglia. Nat Methods 2022; 19:976-985. [PMID: 35879607 DOI: 10.1038/s41592-022-01547-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 06/10/2022] [Indexed: 12/12/2022]
Abstract
As the resident immune cells in the central nervous system (CNS), microglia orchestrate immune responses and dynamically sculpt neural circuits in the CNS. Microglial dysfunction and mutations of microglia-specific genes have been implicated in many diseases of the CNS. Developing effective and safe vehicles for transgene delivery into microglia will facilitate the studies of microglia biology and microglia-associated disease mechanisms. Here, we report the discovery of adeno-associated virus (AAV) variants that mediate efficient in vitro and in vivo microglial transduction via directed evolution of the AAV capsid protein. These AAV-cMG and AAV-MG variants are capable of delivering various genetic payloads into microglia with high efficiency, and enable sufficient transgene expression to support fluorescent labeling, Ca2+ and neurotransmitter imaging and genome editing in microglia in vivo. Furthermore, single-cell RNA sequencing shows that the AAV-MG variants mediate in vivo transgene delivery without inducing microglia immune activation. These AAV variants should facilitate the use of various genetically encoded sensors and effectors in the study of microglia-related biology.
Collapse
|
11
|
Pan X, Yue Y, Boftsi M, Wasala LP, Tran NT, Zhang K, Pintel DJ, Tai PWL, Duan D. Rational engineering of a functional CpG-free ITR for AAV gene therapy. Gene Ther 2022; 29:333-345. [PMID: 34611321 PMCID: PMC8983793 DOI: 10.1038/s41434-021-00296-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
Inverted terminal repeats (ITRs) are the only wild-type components retained in the genome of adeno-associated virus (AAV) vectors. To determine whether ITR modification is a viable approach for AAV vector engineering, we rationally deleted all CpG motifs in the ITR and examined whether CpG elimination compromises AAV-vector production and transduction. Modified ITRs were stable in the plasmid and maintained the CpG-free nature in purified vectors. Replacing the wild-type ITR with the CpG-free ITR did not affect vector genome encapsidation. However, the vector yield was decreased by approximately 3-fold due to reduced vector genome replication. To study the biological potency, we made micro-dystrophin (μDys) AAV vectors carrying either the wild-type ITR or the CpG-free ITR. We delivered the CpG-free μDys vector to one side of the tibialis anterior muscle of dystrophin-null mdx mice and the wild-type μDys vector to the contralateral side. Evaluation at four months after injection showed no difference in the vector genome copy number, microdystrophin expression, and muscle histology and force. Our results suggest that the complete elimination of the CpG motif in the ITR does not affect the biological activity of the AAV vector. CpG-free ITRs could be useful in engineering therapeutic AAV vectors.
Collapse
Affiliation(s)
- Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Maria Boftsi
- Pathobiology Area Graduate Program, University of Missouri, Columbia, MO, 65212, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65212, USA
| | - Lakmini P Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
- Pathobiology Area Graduate Program, University of Missouri, Columbia, MO, 65212, USA
| | - Ngoc Tam Tran
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - David J Pintel
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, 65212, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA.
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65212, USA.
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA.
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
12
|
Wasala NB, Million ED, Watkins TB, Wasala LP, Han J, Yue Y, Lu B, Chen SJ, Hakim CH, Duan D. The gRNA Vector Level Determines the Outcome of Systemic AAV CRISPR Therapy for Duchenne Muscular Dystrophy. Hum Gene Ther 2022; 33:518-528. [PMID: 35350865 PMCID: PMC9142771 DOI: 10.1089/hum.2021.130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 02/13/2022] [Indexed: 01/19/2023] Open
Abstract
Adeno-associated virus (AAV)-mediated clustered regularly interspaced short palindromic repeats (CRISPR) editing holds promise to restore missing dystrophin in Duchenne muscular dystrophy (DMD). Intramuscular coinjection of CRISPR-associated protein 9 (Cas9) and guide RNA (gRNA) vectors resulted in robust dystrophin restoration in short-term studies in the mdx mouse model of DMD. Intriguingly, this strategy failed to yield efficient dystrophin rescue in muscle in a long-term (18-month) systemic injection study. In-depth analyses revealed a selective loss of the gRNA vector after long-term systemic, but not short-term local injection. To determine whether preferential gRNA vector depletion is due to the mode of delivery (local vs. systemic) or the duration of the study (short term vs. long term), we conducted a short-term systemic injection study. The gRNA (4e12 vg/mouse in the 1:1 group or 1.2e13 vg/mouse in the 3:1 group) and Cas9 (4e12 vg/mouse) vectors were coinjected intravenously into 4-week-old mdx mice. The ratio of the gRNA to Cas9 vector genome copy dropped from 1:1 and 3:1 at injection to 0.4:1 and 1:1 at harvest 3 months later, suggesting that the route of administration, rather than the experimental duration, determines preferential gRNA vector loss. Consistent with our long-term systemic injection study, the vector ratio did not influence Cas9 expression. However, the 3:1 group showed significantly higher dystrophin expression and genome editing, better myofiber size distribution, and a more pronounced improvement in muscle function and electrocardiography. Our data suggest that the gRNA vector dose determines the outcome of systemic AAV CRISPR therapy for DMD.
Collapse
Affiliation(s)
- Nalinda B. Wasala
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Emily D. Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Thais B. Watkins
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Lakmini P. Wasala
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Jin Han
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | - Shi-jie Chen
- Department of Physics
- Department of Biochemistry, and
- Institute for Data Science and Informatics, The University of Missouri, Columbia, Missouri, USA
| | - Chady H. Hakim
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, Missouri, USA
- Departments of Neurology, School of Medicine, The University of Missouri, Columbia, Missouri, USA
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, The University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
13
|
Zheng A, Arias EB, Wang H, Kwak SE, Pan X, Duan D, Cartee GD. Exercise-Induced Improvement in Insulin-Stimulated Glucose Uptake by Rat Skeletal Muscle Is Absent in Male AS160-Knockout Rats, Partially Restored by Muscle Expression of Phosphomutated AS160, and Fully Restored by Muscle Expression of Wild-Type AS160. Diabetes 2022; 71:219-232. [PMID: 34753801 PMCID: PMC8914290 DOI: 10.2337/db21-0601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022]
Abstract
One exercise session can elevate insulin-stimulated glucose uptake (ISGU) in skeletal muscle, but the mechanisms remain elusive. Circumstantial evidence suggests a role for Akt substrate of 160 kDa (AS160 or TBC1D4). We used genetic approaches to rigorously test this idea. The initial experiment evaluated the role of AS160 in postexercise increase in ISGU using muscles from male wild-type (WT) and AS160-knockout (KO) rats. The next experiment used AS160-KO rats with an adeno-associated virus (AAV) approach to determine if rescuing muscle AS160 deficiency could restore the ability of exercise to improve ISGU. The third experiment tested if eliminating the muscle GLUT4 deficit in AS160-KO rats via AAV-delivered GLUT4 would enable postexercise enhancement of ISGU. The final experiment used AS160-KO rats and AAV delivery of AS160 mutated to prevent phosphorylation of Ser588, Thr642, and Ser704 to evaluate their role in postexercise ISGU. We discovered the following: 1) AS160 expression was essential for postexercise increase in ISGU; 2) rescuing muscle AS160 expression of AS160-KO rats restored postexercise enhancement of ISGU; 3) restoring GLUT4 expression in AS160-KO muscle did not rescue the postexercise increase in ISGU; and 4) although AS160 phosphorylation on three key sites was not required for postexercise elevation in ISGU, it was essential for the full exercise effect.
Collapse
Affiliation(s)
- Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Institute of Gerontology, University of Michigan, Ann Arbor, MI
- Corresponding author: Gregory D. Cartee,
| |
Collapse
|
14
|
Hakim CH, Kumar SRP, Pérez-López DO, Wasala NB, Zhang D, Yue Y, Teixeira J, Pan X, Zhang K, Million ED, Nelson CE, Metzger S, Han J, Louderman JA, Schmidt F, Feng F, Grimm D, Smith BF, Yao G, Yang NN, Gersbach CA, Chen SJ, Herzog RW, Duan D. Cas9-specific immune responses compromise local and systemic AAV CRISPR therapy in multiple dystrophic canine models. Nat Commun 2021; 12:6769. [PMID: 34819506 PMCID: PMC8613397 DOI: 10.1038/s41467-021-26830-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/21/2021] [Indexed: 11/12/2022] Open
Abstract
Adeno-associated virus (AAV)-mediated CRISPR-Cas9 editing holds promise to treat many diseases. The immune response to bacterial-derived Cas9 has been speculated as a hurdle for AAV-CRISPR therapy. However, immunological consequences of AAV-mediated Cas9 expression have thus far not been thoroughly investigated in large mammals. We evaluate Cas9-specific immune responses in canine models of Duchenne muscular dystrophy (DMD) following intramuscular and intravenous AAV-CRISPR therapy. Treatment results initially in robust dystrophin restoration in affected dogs but also induces muscle inflammation, and Cas9-specific humoral and cytotoxic T-lymphocyte (CTL) responses that are not prevented by the muscle-specific promoter and transient prednisolone immune suppression. In normal dogs, AAV-mediated Cas9 expression induces similar, though milder, immune responses. In contrast, other therapeutic (micro-dystrophin and SERCA2a) and reporter (alkaline phosphatase, AP) vectors result in persistent expression without inducing muscle inflammation. Our results suggest Cas9 immunity may represent a critical barrier for AAV-CRISPR therapy in large mammals.
Collapse
Affiliation(s)
- Chady H Hakim
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Sandeep R P Kumar
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Dennis O Pérez-López
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Dong Zhang
- Department of Physics, The University of Missouri, Columbia, MO, USA
- Department of Biochemistry, The University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, The University of Missouri, Columbia, MO, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - James Teixeira
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Emily D Million
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Christopher E Nelson
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies Biology, Duke University, Durham, NC, USA
| | - Samantha Metzger
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Jin Han
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Jacqueline A Louderman
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Florian Schmidt
- Department of Infectious Diseases/Virology, University of Heidelberg, Heidelberg, Germany
- Cluster of Excellence CellNetworks, University of Heidelberg, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Feng Feng
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, University of Heidelberg, Heidelberg, Germany
- Cluster of Excellence CellNetworks, University of Heidelberg, Heidelberg, Germany
- BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Bruce F Smith
- Department of Pathobiology, Auburn University, Auburn, AL, USA
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, USA
| | - Gang Yao
- Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies Biology, Duke University, Durham, NC, USA
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Shi-Jie Chen
- Department of Physics, The University of Missouri, Columbia, MO, USA
- Department of Biochemistry, The University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, The University of Missouri, Columbia, MO, USA
| | - Roland W Herzog
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO, USA.
- Department of Biomedical, Biological & Chemical Engineering, The University of Missouri, Columbia, MO, USA.
- Department of Neurology, The University of Missouri, Columbia, MO, USA.
- Department of Biomedical Sciences, The University of Missouri, Columbia, MO, USA.
| |
Collapse
|
15
|
Low endotoxin E. coli strain-derived plasmids reduce rAAV vector-mediated immune responses both in vitro and in vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:293-303. [PMID: 34485612 PMCID: PMC8403685 DOI: 10.1016/j.omtm.2021.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/10/2021] [Indexed: 11/24/2022]
Abstract
The major challenge of recombinant adeno-associated virus (rAAV) vectors is host immunological barriers. Compared to the neutralizing antibody and the cytotoxic T lymphocyte response, the host immune responses induced by unsatisfactory rAAV manufacturing were largely ignored previously. rAAV vector production usually requires large amounts of plasmid DNAs. The DNA are commonly isolated from the DH5α bacterial strain, which contains lipopolysaccharide (LPS) contamination. LPS, also named endotoxin, in plasmid DNA is intractable, and residual endotoxin in the subsequent rAAV vectors may result in substantial host immune response. Recently, a ClearColi K12 bacterial strain is commercially available, with genetically modified LPS that does not trigger endotoxic response in mammalian cells. Here, we produced rAAV-DJ vectors by plasmids yielded from either DH5α or ClearColi K12 bacterial strains. Our data indicated that the ClearColi K12 strain had satisfactory protection for the rAAV inverted terminal repeat (ITR) sequence. As expected, the ClearColi K12-derived rAAV-DJ vectors had lower endotoxin levels. The physical and biological equivalency of the purified viral stocks were confirmed by electron micrographs, Coomassie blue staining, and transduction assays. Most importantly, the ClearColi K12-derived rAAV-DJ vectors triggered reduced nuclear factor-kappa B (NF-κB) signaling pathway both in cell cultures in vitro and in C57BL/6 mice retinas in vivo. We believe that the use of the ClearColi K12 bacterial strain could eliminate the LPS in the purified vector stock at the source. Our data indicate its promising use in future clinical development.
Collapse
|
16
|
Takano T, Wallace JT, Baldwin KT, Purkey AM, Uezu A, Courtland JL, Soderblom EJ, Shimogori T, Maness PF, Eroglu C, Soderling SH. Chemico-genetic discovery of astrocytic control of inhibition in vivo. Nature 2020; 588:296-302. [PMID: 33177716 PMCID: PMC8011649 DOI: 10.1038/s41586-020-2926-0] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/19/2020] [Indexed: 01/06/2023]
Abstract
Perisynaptic astrocytic processes are an integral part of central nervous system synapses1,2; however, the molecular mechanisms that govern astrocyte-synapse adhesions and how astrocyte contacts control synapse formation and function are largely unknown. Here we use an in vivo chemico-genetic approach that applies a cell-surface fragment complementation strategy, Split-TurboID, and identify a proteome that is enriched at astrocyte-neuron junctions in vivo, which includes neuronal cell adhesion molecule (NRCAM). We find that NRCAM is expressed in cortical astrocytes, localizes to perisynaptic contacts and is required to restrict neuropil infiltration by astrocytic processes. Furthermore, we show that astrocytic NRCAM interacts transcellularly with neuronal NRCAM coupled to gephyrin at inhibitory postsynapses. Depletion of astrocytic NRCAM reduces numbers of inhibitory synapses without altering glutamatergic synaptic density. Moreover, loss of astrocytic NRCAM markedly decreases inhibitory synaptic function, with minor effects on excitation. Thus, our results present a proteomic framework for how astrocytes interface with neurons and reveal how astrocytes control GABAergic synapse formation and function.
Collapse
Affiliation(s)
- Tetsuya Takano
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA.
| | - John T Wallace
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Katherine T Baldwin
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Alicia M Purkey
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Akiyoshi Uezu
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA
| | - Jamie L Courtland
- Department of Neurobiology, Duke University Medical School, Durham, NC, USA
| | - Erik J Soderblom
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA.,Duke Proteomics and Metabolomics Shared Resource and Duke Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC, USA
| | - Tomomi Shimogori
- Molecular Mechanisms of Brain Development, Center for Brain Science (CBS), RIKEN, Saitama, Japan
| | - Patricia F Maness
- Department of Biochemistry, University of North Carolina School of Medicine, Chapel Hill, NC, USA.,Department of Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Cagla Eroglu
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA. .,Department of Neurobiology, Duke University Medical School, Durham, NC, USA.
| | - Scott H Soderling
- The Department of Cell Biology, Duke University Medical School, Durham, NC, USA. .,Department of Neurobiology, Duke University Medical School, Durham, NC, USA.
| |
Collapse
|
17
|
Hakim CH, Clément N, Wasala LP, Yang HT, Yue Y, Zhang K, Kodippili K, Adamson-Small L, Pan X, Schneider JS, Yang NN, Chamberlain JS, Byrne BJ, Duan D. Micro-dystrophin AAV Vectors Made by Transient Transfection and Herpesvirus System Are Equally Potent in Treating mdx Mouse Muscle Disease. Mol Ther Methods Clin Dev 2020; 18:664-678. [PMID: 32775499 PMCID: PMC7403893 DOI: 10.1016/j.omtm.2020.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/06/2020] [Indexed: 12/26/2022]
Abstract
Vector production scale-up is a major barrier in systemic adeno-associated virus (AAV) gene therapy. Many scalable manufacturing methods have been developed. However, the potency of the vectors generated by these methods has rarely been compared with vectors made by transient transfection (TT), the most commonly used method in preclinical studies. In this study, we blindly compared therapeutic efficacy of an AAV9 micro-dystrophin vector generated by the TT method and scalable herpes simplex virus (HSV) system in a Duchenne muscular dystrophy mouse model. AAV was injected intravenously at 5 × 1014 (high), 5 × 1013 (medium), or 5 × 1012 (low) viral genomes (vg)/kg. Comparable levels of micro-dystrophin expression were observed at each dose in a dose-dependent manner irrespective of the manufacturing method. Vector biodistribution was similar in mice injected with either the TT or the HSV method AAV. Evaluation of muscle degeneration/regeneration showed equivalent protection by vectors made by either method in a dose-dependent manner. Muscle function was similarly improved in a dose-dependent manner irrespective of the vector production method. No apparent toxicity was observed in any mouse. Collectively, our results suggest that the biological potency of the AAV micro-dystrophin vector made by the scalable HSV method is comparable to that made by the TT method.
Collapse
Affiliation(s)
- Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Nathalie Clément
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL, USA
| | - Lakmini P. Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Hsiao T. Yang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Laura Adamson-Small
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | - N. Nora Yang
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Jeffrey S. Chamberlain
- Department of Neurology, Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Barry J. Byrne
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA
| |
Collapse
|
18
|
Shanmugasundaram R, Mortada M, Murugesan GR, Selvaraj RK. In vitro characterization and analysis of probiotic species in the chicken intestine by real-time polymerase chain reaction. Poult Sci 2020; 98:5840-5846. [PMID: 30982074 DOI: 10.3382/ps/pez188] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/27/2019] [Indexed: 12/24/2022] Open
Abstract
Two experiments, 1 in vitro and 1 in vivo study, were conducted to analyze probiotic species characteristics and survival in the intestine of broiler birds. The in vitro study characterized the effect of bile salt supplementation and pH on the proliferation of Lactobacillus reuteri, Pediococcus acidilactici, Bifidobacterium animalis, and Enterococcus faecium. L. reuteri and P. acidilactici growth was maximal when the media was supplemented with 1.0% bile salt, whereas B. animalis and E. faecium growth was maximal when the media was supplemented with 0.5% bile salt. Altering the pH between 2.5 and 5.8 did not significantly (P > 0.05) alter the proliferation of L. reuteri and B. animalis. Decreasing the pH from 5.8 to 2.0 decreased P. acidilactici growth, whereas it increased the E. faecium proliferation. The in vivo study quantified the concentration of L. reuteri, P. acidilactici, B. animalis, E. faecium, and L. salivarius in different intestinal sections from birds supplemented with and without synbiotic containing the above 5 bacteria species. Birds were supplemented with and without synbiotic for 18 d, after which all birds were fed the same basal diet with no synbiotic. At 72 h of feeding, the basal diet with no synbiotics, when the probiotic species in the feed is expected not to confound the recovery of probiotic species from the intestine, intestinal contents were collected. L. reuteri, P. acidilactici, E. faecium, and L. salivarius were below detectable amount in the control group. L. reuteri concentration expressed as copy numbers/g and as percentage of total bacteria was highest in the jejunum and ileum, respectively. E. faecium concentration was highest in the ileum. The copy number of P. acidilactici increased at the duodenum and plateaued after duodenum. L. salivarius concentration was highest in the jejunum. It can be concluded that real-time PCR can be applied to quantify the concentrations of probiotic species in the intestine and probiotic species differ in their ability to colonize different sections of the intestine.
Collapse
Affiliation(s)
- Revathi Shanmugasundaram
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.,BIOMIN America Inc., Overland Park, KS 66210, USA
| | - Mohamad Mortada
- Department of Poultry Sciences, University of Georgia, Athens, GA, 30602, USA
| | | | - Ramesh K Selvaraj
- Department of Poultry Sciences, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
19
|
Wasala NB, Yue Y, Lostal W, Wasala LP, Niranjan N, Hajjar RJ, Babu GJ, Duan D. Single SERCA2a Therapy Ameliorated Dilated Cardiomyopathy for 18 Months in a Mouse Model of Duchenne Muscular Dystrophy. Mol Ther 2020; 28:845-854. [PMID: 31981493 DOI: 10.1016/j.ymthe.2019.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/20/2019] [Accepted: 12/28/2019] [Indexed: 01/16/2023] Open
Abstract
Loss of dystrophin leads to Duchenne muscular dystrophy (DMD). A pathogenic feature of DMD is the significant elevation of cytosolic calcium. Supraphysiological calcium triggers protein degradation, membrane damage, and eventually muscle death and dysfunction. Sarcoplasmic/endoplasmic reticulum (SR) calcium ATPase (SERCA) is a calcium pump that transports cytosolic calcium to the SR during excitation-contraction coupling. We hypothesize that a single systemic delivery of SERCA2a with adeno-associated virus (AAV) may improve calcium recycling and provide long-lasting benefits in DMD. To test this, we injected an AAV9 human SERCA2a vector (6 × 1012 viral genome particles/mouse) intravenously to 3-month-old mdx mice, the most commonly used DMD model. Immunostaining and western blot showed robust human SERCA2a expression in the heart and skeletal muscle for 18 months. Concomitantly, SR calcium uptake was significantly improved in these tissues. SERCA2a therapy significantly enhanced grip force and treadmill performance, completely prevented myocardial fibrosis, and normalized electrocardiograms (ECGs). Cardiac catheterization showed normalization of multiple systolic and diastolic hemodynamic parameters in treated mice. Importantly, chamber dilation was completely prevented, and ejection fraction was restored to the wild-type level. Our results suggest that a single systemic AAV9 SERCA2a therapy has the potential to provide long-lasting benefits for DMD.
Collapse
Affiliation(s)
- Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - William Lostal
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Lakmini P Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Nandita Niranjan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | | | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
20
|
Nance ME, Shi R, Hakim CH, Wasala NB, Yue Y, Pan X, Zhang T, Robinson CA, Duan SX, Yao G, Yang NN, Chen SJ, Wagner KR, Gersbach CA, Duan D. AAV9 Edits Muscle Stem Cells in Normal and Dystrophic Adult Mice. Mol Ther 2019; 27:1568-1585. [PMID: 31327755 PMCID: PMC6731180 DOI: 10.1016/j.ymthe.2019.06.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/27/2022] Open
Abstract
CRISPR editing of muscle stem cells (MuSCs) with adeno-associated virus serotype-9 (AAV9) holds promise for sustained gene repair therapy for muscular dystrophies. However, conflicting evidence exists on whether AAV9 transduces MuSCs. To rigorously address this question, we used a muscle graft model. The grafted muscle underwent complete necrosis before regenerating from its MuSCs. We injected AAV9.Cre into Ai14 mice. These mice express tdTomato upon Cre-mediated removal of a floxed stop codon. About 28%-47% and 24%-89% of Pax7+ MuSCs expressed tdTomato in pre-grafts and regenerated grafts (p > 0.05), respectively, suggesting AAV9 efficiently transduced MuSCs, and AAV9-edited MuSCs renewed successfully. Robust MuSC transduction was further confirmed by delivering AAV9.Cre to Pax7-ZsGreen-Ai14 mice in which Pax7+ MuSCs are genetically labeled by ZsGreen. Next, we co-injected AAV9.Cas9 and AAV9.gRNA to dystrophic mdx mice to repair the mutated dystrophin gene. CRISPR-treated and untreated muscles were grafted to immune-deficient, dystrophin-null NSG.mdx4cv mice. Grafts regenerated from CRISPR-treated muscle contained the edited genome and yielded 2.7-fold more dystrophin+ cells (p = 0.015). Importantly, increased dystrophin expression was not due to enhanced formation of revertant fibers or de novo transduction by residual CRISPR vectors in the graft. We conclude that AAV9 effectively transduces MuSCs. AAV9 CRISPR editing of MuSCs may provide enduring therapy.
Collapse
MESH Headings
- Animals
- Clustered Regularly Interspaced Short Palindromic Repeats
- Dependovirus/genetics
- Disease Models, Animal
- Dystrophin/chemistry
- Dystrophin/genetics
- Gene Editing
- Gene Expression
- Gene Transfer Techniques
- Genes, Reporter
- Genetic Vectors/genetics
- Mice
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/therapy
- Myoblasts/metabolism
- RNA, Guide, CRISPR-Cas Systems/genetics
- Regeneration
- Satellite Cells, Skeletal Muscle/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- Michael E Nance
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Ruicheng Shi
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, USA
| | - Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Tracy Zhang
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD 21205, USA; Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Carolyn A Robinson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Sean X Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Gang Yao
- Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA
| | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, USA
| | - Shi-Jie Chen
- Department of Physics, University of Missouri, Columbia, MO 65212, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD 21205, USA; Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
21
|
Pan X, Sands SA, Yue Y, Zhang K, LeVine SM, Duan D. An Engineered Galactosylceramidase Construct Improves AAV Gene Therapy for Krabbe Disease in Twitcher Mice. Hum Gene Ther 2019; 30:1039-1051. [PMID: 31184217 PMCID: PMC6761594 DOI: 10.1089/hum.2019.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/16/2019] [Indexed: 12/30/2022] Open
Abstract
Krabbe disease is an inherited neurodegenerative disease caused by mutations in the galactosylceramidase gene. In the infantile form, patients die before 3 years of age. Systemic adeno-associated virus serotype 9 (AAV9) gene therapy was recently shown to reverse the disease course in human patients in another lethal infantile neurodegenerative disease. To explore AAV9 therapy for Krabbe disease, we engineered a codon-optimized AAV9 galactosylceramidase vector. We further incorporated features to allow AAV9-derived galactosylceramidase to more efficiently cross the blood-brain barrier and be secreted from transduced cells. We tested the optimized vector by a single systemic injection in the twitcher mouse, an authentic Krabbe disease model. Untreated twitcher mice showed characteristic neuropathology and motion defects. They died prematurely with a median life span of 41 days. Intravenous injection in 2-day-old twitcher mice reduced central and peripheral neuropathology and significantly improved the gait pattern and body weight. Noticeably, the median life span was extended to 150 days. Intraperitoneal injection in 6- to 12-day-old twitcher mice also significantly improved the motor function, body weight, and median life span (to 104 days). Our results far exceed the ≤70 days median life span seen in all reported stand-alone systemic AAV therapies. Our study highlights the importance of vector engineering for Krabbe disease gene therapy. The engineered vector warrants further development.
Collapse
Affiliation(s)
- Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Scott A. Sands
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Steven M. LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri
| |
Collapse
|
22
|
Man A, Slevin M, Petcu E, Fraefel C. The Cyclin-Dependent Kinase 5 Inhibitor Peptide Inhibits Herpes Simplex Virus Type 1 Replication. Sci Rep 2019; 9:1260. [PMID: 30718749 PMCID: PMC6362106 DOI: 10.1038/s41598-018-37989-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/18/2018] [Indexed: 11/09/2022] Open
Abstract
In order to evaluate the influence of CDK5 inhibitory peptide (CIP) on Human alphaherpesvirus 1 (HSV-1) replication, we constructed two recombinant adeno-associated-virus 2 (rAAV2) vectors encoding CIP fused with cyan-fluorescent-protein (CFP), with or without nuclear localization signal. A third vector encoding non-fused CIP and CFP was also constructed. HeLa and HEK 293T cells were infected with the rAAV-CIP vectors at multiplicity of infection (MOI) of 5000, in the absence or presence of a recombinant HSV-1 that encodes a yellow-fluorescent-protein (rHSV48Y; MOI = 1). Cells co-infected with rHSV48Y and rAAV vectors that did not express the CIP gene (rAAV-CFP-Neo) served as controls. At 24 h after infection, the effect of CIP on rHSV48Y replication was assessed by PCR, qRT-PCR, Western-blot, flow-cytometry, epifluorescence and confocal microscopy. We show that in cultures co-infected with rAAV-CFP-Neo, 27% of the CFP-positive cells present rHSV48Y replication compartments. By contrast, in cultures co-infected with CIP-encoding rAAV2 vectors and rHSV48Y only 6-20% of the cells positive for CIP showed rHSV48Y replication compartments, depending on the CIP variant. Flow-cytometry showed that less than 40% of the rHSV48Y/rAAV-CIP, and more than 75% of rHSV48Y/rAAV-CFP-Neo co-infected cells were positive for both transgene products. The microscopy and flow-cytometry data support the hypothesis that CIP is inhibiting HSV-1 replication.
Collapse
Affiliation(s)
- Adrian Man
- Institute of Virology, University of Zurich, Zurich, Switzerland
- Department of Microbiology, University of Medicine and Pharmacy of Tîrgu Mureș, Târgu Mureș, Romania
| | - Mark Slevin
- University of Medicine and Pharmacy of Tîrgu Mureș, Târgu Mureș, Romania.
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK.
| | - Eugen Petcu
- Griffith University, Gold Coast, Brisbane, Australia
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Hakim CH, Wasala NB, Nelson CE, Wasala LP, Yue Y, Louderman JA, Lessa TB, Dai A, Zhang K, Jenkins GJ, Nance ME, Pan X, Kodippili K, Yang NN, Chen SJ, Gersbach CA, Duan D. AAV CRISPR editing rescues cardiac and muscle function for 18 months in dystrophic mice. JCI Insight 2018; 3:124297. [PMID: 30518686 PMCID: PMC6328021 DOI: 10.1172/jci.insight.124297] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/31/2018] [Indexed: 01/25/2023] Open
Abstract
Adeno-associated virus-mediated (AAV-mediated) CRISPR editing is a revolutionary approach for treating inherited diseases. Sustained, often life-long mutation correction is required for treating these diseases. Unfortunately, this has never been demonstrated with AAV CRISPR therapy. We addressed this question in the mdx model of Duchenne muscular dystrophy (DMD). DMD is caused by dystrophin gene mutation. Dystrophin deficiency leads to ambulation loss and cardiomyopathy. We treated 6-week-old mice intravenously and evaluated disease rescue at 18 months. Surprisingly, nominal dystrophin was restored in skeletal muscle. Cardiac dystrophin was restored, but histology and hemodynamics were not improved. To determine the underlying mechanism, we evaluated components of the CRISPR-editing machinery. Intriguingly, we found disproportional guide RNA (gRNA) vector depletion. To test whether this is responsible for the poor outcome, we increased the gRNA vector dose and repeated the study. This strategy significantly increased dystrophin restoration and reduced fibrosis in all striated muscles at 18 months. Importantly, skeletal muscle function and cardiac hemodynamics were significantly enhanced. Interestingly, we did not see selective depletion of the gRNA vector after intramuscular injection. Our results suggest that gRNA vector loss is a unique barrier for systemic AAV CRISPR therapy. This can be circumvented by vector dose optimization.
Collapse
Affiliation(s)
- Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- National Center for Advancing Translational Sciences, NIH, Rockville, Maryland, USA
| | - Nalinda B. Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Christopher E. Nelson
- Department of Biomedical Engineering and
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
| | - Lakmini P. Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Jacqueline A. Louderman
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Thais B. Lessa
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Aihua Dai
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Gregory J. Jenkins
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Michael E. Nance
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - N. Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, Maryland, USA
| | - Shi-jie Chen
- Department of Physics and
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering and
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Neurology, School of Medicine
- Department of Bioengineering, and
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
24
|
Lin R, Wang R, Yuan J, Feng Q, Zhou Y, Zeng S, Ren M, Jiang S, Ni H, Zhou C, Gong H, Luo M. Cell-type-specific and projection-specific brain-wide reconstruction of single neurons. Nat Methods 2018; 15:1033-1036. [PMID: 30455464 DOI: 10.1038/s41592-018-0184-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/31/2018] [Indexed: 11/09/2022]
Abstract
We developed a dual-adeno-associated-virus expression system that enables strong and sparse labeling of individual neurons with cell-type and projection specificity. We demonstrated its utility for whole-brain reconstruction of midbrain dopamine neurons and striatum-projecting cortical neurons. We further extended the labeling method for rapid reconstruction in cleared thick brain sections and simultaneous dual-color labeling. This labeling system may facilitate the process of generating mesoscale single-neuron projectomes of mammalian brains.
Collapse
Affiliation(s)
- Rui Lin
- School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Ruiyu Wang
- School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China
| | - Qiru Feng
- National Institute of Biological Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Youtong Zhou
- National Institute of Biological Sciences, Beijing, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Miao Ren
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Jiang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Ni
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Can Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China. .,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China. .,HUST-Suzhou Institute for Brainsmatics, JITRI Institute for Brainsmatics, Suzhou, China.
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China. .,School of Life Sciences, Tsinghua University, Beijing, China. .,Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
25
|
Heider S, Muzard J, Zaruba M, Metzner C. Integrated Method for Purification and Single-Particle Characterization of Lentiviral Vector Systems by Size Exclusion Chromatography and Tunable Resistive Pulse Sensing. Mol Biotechnol 2018; 59:251-259. [PMID: 28567687 PMCID: PMC5486506 DOI: 10.1007/s12033-017-0009-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elements derived from lentiviral particles such as viral vectors or virus-like particles are commonly used for biotechnological and biomedical applications, for example in mammalian protein expression, gene delivery or therapy, and vaccine development. Preparations of high purity are necessary in most cases, especially for clinical applications. For purification, a wide range of methods are available, from density gradient centrifugation to affinity chromatography. In this study we have employed size exclusion columns specifically designed for the easy purification of extracellular vesicles including exosomes. In addition to viral marker protein and total protein analysis, a well-established single-particle characterization technology, termed tunable resistive pulse sensing, was employed to analyze fractions of highest particle load and purity and characterize the preparations by size and surface charge/electrophoretic mobility. With this study, we propose an integrated platform combining size exclusion chromatography and tunable resistive pulse sensing for monitoring production and purification of viral particles.
Collapse
Affiliation(s)
- Susanne Heider
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.,Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Julien Muzard
- Izon Science, 8C Homersham Place, PO Box 39168, Burnside, Christchurch, 8053, New Zealand
| | - Marianne Zaruba
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Christoph Metzner
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
26
|
Kennedy LH, Rinholm JE. Visualization and Live Imaging of Oligodendrocyte Organelles in Organotypic Brain Slices Using Adeno-associated Virus and Confocal Microscopy. J Vis Exp 2017:56237. [PMID: 29155726 PMCID: PMC5755187 DOI: 10.3791/56237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Neurons rely on the electric insulation and trophic support of myelinating oligodendrocytes. Despite the importance of oligodendrocytes, the advanced tools currently used to study neurons, have only partly been taken on by oligodendrocyte researchers. Cell type-specific staining by viral transduction is a useful approach to study live organelle dynamics. This paper describes a protocol for visualizing oligodendrocyte mitochondria in organotypic brain slices by transduction with adeno-associated virus (AAV) carrying genes for mitochondrial targeted fluorescent proteins under the transcriptional control of the myelin basic protein promoter. It includes the protocol for making organotypic coronal mouse brain slices. A procedure for time-lapse imaging of mitochondria then follows. These methods can be transferred to other organelles and may be particularly useful for studying organelles in the myelin sheath. Finally, we describe a readily available technique for visualization of unstained myelin in living slices by Confocal Reflectance microscopy (CoRe). CoRe requires no extra equipment and can be useful to identify the myelin sheath during live imaging.
Collapse
Affiliation(s)
| | - Johanne Egge Rinholm
- Division of Anatomy, Institute of Basic Medical Sciences, University of Oslo; Department of Microbiology, Oslo University Hospital;
| |
Collapse
|
27
|
Kodippili K, Hakim CH, Pan X, Yang HT, Yue Y, Zhang Y, Shin JH, Yang NN, Duan D. Dual AAV Gene Therapy for Duchenne Muscular Dystrophy with a 7-kb Mini-Dystrophin Gene in the Canine Model. Hum Gene Ther 2017; 29:299-311. [PMID: 28793798 DOI: 10.1089/hum.2017.095] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Dual adeno-associated virus (AAV) technology was developed in 2000 to double the packaging capacity of the AAV vector. The proof of principle has been demonstrated in various mouse models. Yet, pivotal evidence is lacking in large animal models of human diseases. Here we report expression of a 7-kb canine ΔH2-R15 mini-dystrophin gene using a pair of dual AAV vectors in the canine model of Duchenne muscular dystrophy (DMD). The ΔH2-R15 minigene is by far the most potent synthetic dystrophin gene engineered for DMD gene therapy. We packaged minigene dual vectors in Y731F tyrosine-modified AAV-9 and delivered to the extensor carpi ulnaris muscle of a 12-month-old affected dog at the dose of 2 × 1013 viral genome particles/vector/muscle. Widespread mini-dystrophin expression was observed 2 months after gene transfer. The missing dystrophin-associated glycoprotein complex was restored. Treatment also reduced muscle degeneration and fibrosis and improved myofiber size distribution. Importantly, dual AAV therapy greatly protected the muscle from eccentric contraction-induced force loss. Our data provide the first clear evidence that dual AAV therapy can be translated to a diseased large mammal. Further development of dual AAV technology may lead to effective therapies for DMD and many other diseases in human patients.
Collapse
Affiliation(s)
- Kasun Kodippili
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri
| | - Chady H Hakim
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri.,2 National Center for Advancing Translational Sciences , Bethesda, Maryland
| | - Xiufang Pan
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri
| | - Hsiao T Yang
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri.,3 Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri , Columbia, Missouri
| | - Yongping Yue
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri
| | - Yadong Zhang
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri
| | - Jin-Hong Shin
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri
| | - N Nora Yang
- 2 National Center for Advancing Translational Sciences , Bethesda, Maryland
| | - Dongsheng Duan
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, Missouri.,3 Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri , Columbia, Missouri.,4 Department of Neurology, School of Medicine, The University of Missouri , Columbia, Missouri.,5 Department of Bioengineering, The University of Missouri , Columbia, Missouri
| |
Collapse
|
28
|
Robert MA, Chahal PS, Audy A, Kamen A, Gilbert R, Gaillet B. Manufacturing of recombinant adeno-associated viruses using mammalian expression platforms. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201600193] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/08/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Marc-André Robert
- Département de génie chimique; Université Laval; Québec QC Canada
- National Research Council Canada; Montréal QC Canada
| | | | - Alexandre Audy
- Département de génie chimique; Université Laval; Québec QC Canada
- National Research Council Canada; Montréal QC Canada
| | - Amine Kamen
- Department of Bioengineering; McGill University; Montréal QC Canada
| | | | - Bruno Gaillet
- Département de génie chimique; Université Laval; Québec QC Canada
| |
Collapse
|
29
|
Zhao J, Kodippili K, Yue Y, Hakim CH, Wasala L, Pan X, Zhang K, Yang NN, Duan D, Lai Y. Dystrophin contains multiple independent membrane-binding domains. Hum Mol Genet 2016; 25:3647-3653. [PMID: 27378693 DOI: 10.1093/hmg/ddw210] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 06/14/2016] [Accepted: 06/27/2016] [Indexed: 01/09/2023] Open
Abstract
Dystrophin is a large sub-sarcolemmal protein. Its absence leads to Duchenne muscular dystrophy (DMD). Binding to the sarcolemma is essential for dystrophin to protect muscle from contraction-induced injury. It has long been thought that membrane binding of dystrophin depends on its cysteine-rich (CR) domain. Here, we provide in vivo evidence suggesting that dystrophin contains three additional membrane-binding domains including spectrin-like repeats (R)1-3, R10-12 and C-terminus (CT). To systematically study dystrophin membrane binding, we split full-length dystrophin into ten fragments and examined subcellular localizations of each fragment by adeno-associated virus-mediated gene transfer. In skeletal muscle, R1-3, CR domain and CT were exclusively localized at the sarcolemma. R10-12 showed both cytosolic and sarcolemmal localization. Importantly, the CR-independent membrane binding was conserved in murine and canine muscles. A critical function of the CR-mediated membrane interaction is the assembly of the dystrophin-associated glycoprotein complex (DGC). While R1-3 and R10-12 did not restore the DGC, surprisingly, CT alone was sufficient to establish the DGC at the sarcolemma. Additional studies suggest that R1-3 and CT also bind to the sarcolemma in the heart, though relatively weak. Taken together, our study provides the first conclusive in vivo evidence that dystrophin contains multiple independent membrane-binding domains. These structurally and functionally distinctive membrane-binding domains provide a molecular framework for dystrophin to function as a shock absorber and signaling hub. Our results not only shed critical light on dystrophin biology and DMD pathogenesis, but also provide a foundation for rationally engineering minimized dystrophins for DMD gene therapy.
Collapse
Affiliation(s)
- Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine.,National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Lakmini Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Nora N Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine .,Department of Neurology, School of Medicine.,Department of Bioengineering.,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine
| |
Collapse
|
30
|
Wasala NB, Lai Y, Shin JH, Zhao J, Yue Y, Duan D. Genomic removal of a therapeutic mini-dystrophin gene from adult mice elicits a Duchenne muscular dystrophy-like phenotype. Hum Mol Genet 2016; 25:2633-2644. [PMID: 27106099 DOI: 10.1093/hmg/ddw123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/29/2016] [Accepted: 04/18/2016] [Indexed: 12/27/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by dystrophin deficiency. A fundamental question in DMD pathogenesis and dystrophin gene therapy is whether muscle health depends on continuous dystrophin expression throughout the life. Published data suggest that transient dystrophin expression in early life might offer permanent protection. To study the consequences of adulthood dystrophin loss, we generated two strains of floxed mini-dystrophin transgenic mice on the dystrophin-null background. Muscle diseases were prevented in skeletal muscle of the YL238 strain and the heart of the SJ13 strain by selective expression of a therapeutic mini-dystrophin gene in skeletal muscle and heart, respectively. The mini-dystrophin gene was removed from the tibialis anterior (TA) muscle of 8-month-old YL238 mice and the heart of 7-month-old SJ13 mice using an adeno-associated virus serotype-9 Cre recombinase vector (AAV.CBA.Cre). At 12 and 15 months after AAV.CBA.Cre injection, mini-dystrophin expression was reduced by ∼87% in the TA muscle of YL238 mice and ∼64% in the heart of SJ13 mice. Mini-dystrophin reduction caused muscle atrophy, degeneration and force loss in the TA muscle of YL238 mice and significantly compromised left ventricular hemodynamics in SJ13 mice. Our results suggest that persistent dystrophin expression is essential for continuous muscle and heart protection.
Collapse
Affiliation(s)
- Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Yi Lai
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Jin-Hong Shin
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Junling Zhao
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine .,Department of Neurology, School of Medicine.,Department of Bioengineering, The University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
31
|
Yue Y, Pan X, Hakim CH, Kodippili K, Zhang K, Shin JH, Yang HT, McDonald T, Duan D. Safe and bodywide muscle transduction in young adult Duchenne muscular dystrophy dogs with adeno-associated virus. Hum Mol Genet 2015; 24:5880-90. [PMID: 26264580 DOI: 10.1093/hmg/ddv310] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/28/2015] [Indexed: 12/17/2022] Open
Abstract
The ultimate goal of muscular dystrophy gene therapy is to treat all muscles in the body. Global gene delivery was demonstrated in dystrophic mice more than a decade ago using adeno-associated virus (AAV). However, translation to affected large mammals has been challenging. The only reported attempt was performed in newborn Duchenne muscular dystrophy (DMD) dogs. Unfortunately, AAV injection resulted in growth delay, muscle atrophy and contracture. Here we report safe and bodywide AAV delivery in juvenile DMD dogs. Three ∼2-m-old affected dogs received intravenous injection of a tyrosine-engineered AAV-9 reporter or micro-dystrophin (μDys) vector at the doses of 1.92-6.24 × 10(14) viral genome particles/kg under transient or sustained immune suppression. DMD dogs tolerated injection well and their growth was not altered. Hematology and blood biochemistry were unremarkable. No adverse reactions were observed. Widespread muscle transduction was seen in skeletal muscle, the diaphragm and heart for at least 4 months (the end of the study). Nominal expression was detected in internal organs. Improvement in muscle histology was observed in μDys-treated dogs. In summary, systemic AAV gene transfer is safe and efficient in young adult dystrophic large mammals. This may translate to bodywide gene therapy in pediatric patients in the future.
Collapse
Affiliation(s)
- Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Jin-Hong Shin
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Hsiao T Yang
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO, USA
| | - Thomas McDonald
- Department of Molecular Microbiology and Immunology, School of Medicine
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, Department of Neurology and
| |
Collapse
|
32
|
Pan X, Yue Y, Zhang K, Hakim CH, Kodippili K, McDonald T, Duan D. AAV-8 is more efficient than AAV-9 in transducing neonatal dog heart. Hum Gene Ther Methods 2015; 26:54-61. [PMID: 25763686 DOI: 10.1089/hgtb.2014.128] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adeno-associated virus serotype-8 and 9 (AAV-8 and 9) are the leading candidate vectors to test bodywide neonatal muscle gene therapy in large mammals. We have previously shown that systemic injection of 2-2.5×10(14) viral genome (vg) particles/kg of AAV-9 resulted in widespread skeletal muscle gene transfer in newborn dogs. However, nominal transduction was observed in the heart. In contrast, robust expression was achieved in both skeletal muscle and heart in neonatal dogs with 7.14-9.06×10(14) vg particles/kg of AAV-8. To determine whether superior cardiac transduction of AAV-8 is because of the higher vector dose, we delivered 6.14×10(14) and 9.65×10(14) vg particles/kg of AAV-9 to newborn puppies via the jugular vein. Transduction was examined 2.5 months later. Consistent with our previous reports, we observed robust bodywide transduction in skeletal muscle. However, increased AAV dose only moderately improved heart transduction. It never reached the level achieved by AAV-8. Our results suggest that differential cardiac transduction by AAV-8 and AAV-9 is likely because of the intrinsic property of the viral capsid rather than the vector dose.
Collapse
Affiliation(s)
- Xiufang Pan
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri , Columbia, MO 65212
| | | | | | | | | | | | | |
Collapse
|
33
|
Lostal W, Kodippili K, Yue Y, Duan D. Full-length dystrophin reconstitution with adeno-associated viral vectors. Hum Gene Ther 2014; 25:552-62. [PMID: 24580018 DOI: 10.1089/hum.2013.210] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common lethal muscle disorder in children. It is caused by mutations of the dystrophin gene. Adeno-associated virus (AAV)-mediated gene replacement therapy has been actively pursued to treat DMD. However, this promising therapeutic modality has been challenged by the small packaging capacity of the AAV vector. The size of the full-length dystrophin cDNA is >11 kb, while an AAV virus can carry only a 5 kb genome. Innovative high-capacity AAV vectors may offer an opportunity to express the full-length dystrophin coding sequence. Here we describe several sets of tri-AAV vectors for full-length human dystrophin delivery. In each set, the full-length human dystrophin cDNA was split into three fragments and independently packaged into separate recombinant AAV vectors. Each vector was engineered with unique recombination signals for directional recombination. Tri-AAV vectors were coinjected into the tibialis anterior muscle of dystrophin-deficient mdx4cv mice. Thirty-five days after injection, dystrophin expression was examined by immunofluorescence staining. Despite low reconstitution efficiency, full-length human dystrophin was successfully expressed from the tri-AAV vectors. Our results suggest that AAV can be engineered to express an extra-large (up to 15 kb) gene that is approximately three times the size of the wild-type AAV genome. Further optimization of the trivector strategy may expand the utility of AAV for human gene therapy.
Collapse
Affiliation(s)
- William Lostal
- 1 Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri , Columbia, MO 65212
| | | | | | | |
Collapse
|
34
|
Systemic gene transfer reveals distinctive muscle transduction profile of tyrosine mutant AAV-1, -6, and -9 in neonatal dogs. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14002. [PMID: 25105153 PMCID: PMC4121663 DOI: 10.1038/mtm.2014.2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The muscular dystrophies are a group of devastating genetic disorders that affect both skeletal and cardiac muscle. An effective gene therapy for these diseases requires bodywide muscle delivery. Tyrosine mutant adeno-associated virus (AAV) has been considered as a class of highly potent gene transfer vectors. Here, we tested the hypothesis that systemic delivery of tyrosine mutant AAV can result in bodywide muscle transduction in newborn dogs. Three tyrosine mutant AAV vectors (Y445F/Y731F AAV-1, Y445F AAV-6, and Y731F AAV-9) were evaluated. These vectors expressed the alkaline phosphatase reporter gene under transcriptional regulation of either the muscle-specific Spc5-12 promoter or the ubiquitous Rous sarcoma virus promoter. Robust skeletal and cardiac muscle transduction was achieved with Y445F/Y731F AAV-1. However, Y731F AAV-9 only transduced skeletal muscle. Surprisingly, Y445F AAV-6 resulted in minimal muscle transduction. Serological study suggests that the preexisting neutralization antibody may underlie the limited transduction of Y445F AAV-6. In summary, we have identified Y445F/Y731F AAV-1 as a potentially excellent systemic gene transfer vehicle to target both skeletal muscle and the heart in neonatal puppies. Our findings have important implications in exploring systemic neonatal gene therapy in canine models of muscular dystrophy.
Collapse
|
35
|
Pan X, Yue Y, Zhang K, Lostal W, Shin JH, Duan D. Long-term robust myocardial transduction of the dog heart from a peripheral vein by adeno-associated virus serotype-8. Hum Gene Ther 2013; 24:584-94. [PMID: 23551085 PMCID: PMC3689160 DOI: 10.1089/hum.2013.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/01/2013] [Indexed: 12/31/2022] Open
Abstract
Molecular intervention using noninvasive myocardial gene transfer holds great promise for treating heart diseases. Robust cardiac transduction from peripheral vein injection has been achieved in rodents using adeno-associated virus (AAV) serotype-9 (AAV-9). However, a similar approach has failed to transduce the heart in dogs, a commonly used large animal model for heart diseases. To develop an effective noninvasive method to deliver exogenous genes to the dog heart, we employed an AAV-8 vector that expresses human placental alkaline phosphatase reporter gene under the transcriptional regulation of the Rous sarcoma virus promoter. Vectors were delivered to three neonatal dogs at the doses of 1.35×10(14), 7.14×10(14), and 9.06×10(14) viral genome particles/kg body weight via the jugular vein. Transduction efficiency and overall safety were evaluated at 1.5, 2.5, and 12 months postinjection. AAV delivery was well tolerated and dog growth was normal. Blood chemistry and internal organ histology were unremarkable. Widespread skeletal muscle transduction was observed in all dogs without T-cell infiltration. Encouragingly, whole heart myocardial transduction was achieved in two dogs that received higher doses and cardiac expression lasted for at least 1 year. In summary, peripheral vein AAV-8 injection may represent a simple heart gene transfer method in large mammals. Further optimization of this gene delivery strategy may open the door for a readily applicable gene therapy method to treat many heart diseases.
Collapse
Affiliation(s)
- Xiufang Pan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
| | - William Lostal
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
- Current address: Génethon, 91002 Evry Cedex, France
| | - Jin-Hong Shin
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
- Current address: Pusan National University Yangsan Hospital, Yangsan 626-770, Republic of Korea
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO 65212
| |
Collapse
|
36
|
Zhang Y, Yue Y, Li L, Hakim CH, Zhang K, Thomas GD, Duan D. Dual AAV therapy ameliorates exercise-induced muscle injury and functional ischemia in murine models of Duchenne muscular dystrophy. Hum Mol Genet 2013; 22:3720-9. [PMID: 23681067 DOI: 10.1093/hmg/ddt224] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neuronal nitric oxide synthase (nNOS) membrane delocalization contributes to the pathogenesis of Duchenne muscular dystrophy (DMD) by promoting functional muscle ischemia and exacerbating muscle injury during exercise. We have previously shown that supra-physiological expression of nNOS-binding mini-dystrophin restores normal blood flow regulation and prevents functional ischemia in transgenic mdx mice, a DMD model. A critical next issue is whether systemic dual adeno-associated virus (AAV) gene therapy can restore nNOS-binding mini-dystrophin expression and mitigate muscle activity-related functional ischemia and injury. Here, we performed systemic gene transfer in mdx and mdx4cv mice using a pair of dual AAV vectors that expressed a 6 kb nNOS-binding mini-dystrophin gene. Vectors were packaged in tyrosine mutant AAV-9 and co-injected (5 × 10(12) viral genome particles/vector/mouse) via the tail vein to 1-month-old dystrophin-null mice. Four months later, we observed 30-50% mini-dystrophin positive myofibers in limb muscles. Treatment ameliorated histopathology, increased muscle force and protected against eccentric contraction-induced injury. Importantly, dual AAV therapy successfully prevented chronic exercise-induced muscle force drop. Doppler hemodynamic assay further showed that therapy attenuated adrenergic vasoconstriction in contracting muscle. Our results suggest that partial transduction can still ameliorate nNOS delocalization-associated functional deficiency. Further evaluation of nNOS binding mini-dystrophin dual AAV vectors is warranted in dystrophic dogs and eventually in human patients.
Collapse
Affiliation(s)
- Yadong Zhang
- Department of Molecular Microbiology and Immunology, The University of Missouri, Columbia, MO 65212, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Microdystrophin ameliorates muscular dystrophy in the canine model of duchenne muscular dystrophy. Mol Ther 2013; 21:750-7. [PMID: 23319056 DOI: 10.1038/mt.2012.283] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Dystrophin deficiency results in lethal Duchenne muscular dystrophy (DMD). Substituting missing dystrophin with abbreviated microdystrophin has dramatically alleviated disease in mouse DMD models. Unfortunately, translation of microdystrophin therapy has been unsuccessful in dystrophic dogs, the only large mammalian model. Approximately 70% of the dystrophin-coding sequence is removed in microdystrophin. Intriguingly, loss of ≥50% dystrophin frequently results in severe disease in patients. To test whether the small gene size constitutes a fundamental design error for large mammalian muscle, we performed a comprehensive study using 22 dogs (8 normal and 14 dystrophic). We delivered the ΔR2-15/ΔR18-19/ΔR20-23/ΔC microdystrophin gene to eight extensor carpi ulnaris (ECU) muscles in six dystrophic dogs using Y713F tyrosine mutant adeno-associated virus (AAV)-9 (2.6 × 10(13) viral genome (vg) particles/muscle). Robust expression was observed 2 months later despite T-cell infiltration. Major components of the dystrophin-associated glycoprotein complex (DGC) were restored by microdystrophin. Treated muscle showed less inflammation, fibrosis, and calcification. Importantly, therapy significantly preserved muscle force under the stress of repeated cycles of eccentric contraction. Our results have established the proof-of-concept for microdystrophin therapy in dystrophic muscles of large mammals and set the stage for clinical trial in human patients.
Collapse
|