1
|
Loeb EJ, Havlik PL, Elmore ZC, Rosales A, Fergione SM, Gonzalez TJ, Smith TJ, Benkert AR, Fiflis DN, Asokan A. Capsid-mediated control of adeno-associated viral transcription determines host range. Cell Rep 2024; 43:113902. [PMID: 38431840 PMCID: PMC11150003 DOI: 10.1016/j.celrep.2024.113902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/13/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Adeno-associated virus (AAV) is a member of the genus Dependoparvovirus, which infects a wide range of vertebrate species. Here, we observe that, unlike most primate AAV isolates, avian AAV is transcriptionally silenced in human cells. By swapping the VP1 N terminus from primate AAVs (e.g., AAV8) onto non-mammalian isolates (e.g., avian AAV), we identify a minimal component of the AAV capsid that controls viral transcription and unlocks robust transduction in both human cells and mouse tissue. This effect is accompanied by increased AAV genome chromatin accessibility and altered histone methylation. Proximity ligation analysis reveals that host factors are selectively recruited by the VP1 N terminus of AAV8 but not avian AAV. Notably, these include AAV essential factors implicated in the nuclear factor κB pathway, chromatin condensation, and histone methylation. We postulate that the AAV capsid has evolved mechanisms to recruit host factors to its genome, allowing transcriptional activation in a species-specific manner.
Collapse
Affiliation(s)
- Ezra J Loeb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Patrick L Havlik
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Zachary C Elmore
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Alan Rosales
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sophia M Fergione
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Trevor J Gonzalez
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Timothy J Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Abigail R Benkert
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - David N Fiflis
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Aravind Asokan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
2
|
El Haddad L, Lai E, Murthy PKL, Biswas DD, Soufny R, Roger AL, Tata PR, ElMallah MK. GAA deficiency disrupts distal airway cells in Pompe disease. Am J Physiol Lung Cell Mol Physiol 2023; 325:L288-L298. [PMID: 37366541 PMCID: PMC10625827 DOI: 10.1152/ajplung.00032.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023] Open
Abstract
Pompe disease is an autosomal recessive glycogen storage disease caused by mutations in the gene that encodes acid alpha-glucosidase (GAA)-an enzyme responsible for hydrolyzing lysosomal glycogen. GAA deficiency results in systemic lysosomal glycogen accumulation and cellular disruption. Glycogen accumulation in skeletal muscles, motor neurons, and airway smooth muscle cells is known to contribute to respiratory insufficiency in Pompe disease. However, the impact of GAA deficiency on the distal alveolar type 1 and type 2 cells (AT1 and AT2) has not been evaluated. AT1 cells rely on lysosomes for cellular homeostasis so that they can maintain a thin barrier for gas exchange, whereas AT2 cells depend on lysosome-like structures (lamellar bodies) for surfactant production. Using a mouse model of Pompe disease, the Gaa-/- mouse, we investigated the consequences of GAA deficiency on AT1 and AT2 cells using histology, pulmonary function and mechanics, and transcriptional analysis. Histological analysis revealed increased accumulation of lysosomal-associated membrane protein 1 (LAMP1) in the Gaa-/- mice lungs. Furthermore, ultrastructural examination showed extensive intracytoplasmic vacuoles enlargement and lamellar body engorgement. Respiratory dysfunction was confirmed using whole body plethysmography and forced oscillometry. Finally, transcriptomic analysis demonstrated dysregulation of surfactant proteins in AT2 cells, specifically reduced levels of surfactant protein D in the Gaa-/- mice. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the distal airway cells that disrupts surfactant homeostasis and contributes to respiratory impairments in Pompe disease.NEW & NOTEWORTHY This research highlights the impact of Pompe disease on distal airway cells. Prior to this work, respiratory insufficiency in Pompe disease was classically attributed to pathology in respiratory muscles and motor neurons. Using the Pompe mouse model, we note significant pathology in alveolar type 1 and 2 cells with reductions in surfactant protein D and disrupted surfactant homeostasis. These novel findings highlight the potential contributions of alveolar pathology to respiratory insufficiency in Pompe disease.
Collapse
Affiliation(s)
- Léa El Haddad
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| | - Elias Lai
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| | | | - Debolina D Biswas
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| | - Rania Soufny
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| | - Angela L Roger
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| | | | - Mai K ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, North Carolina, United States
| |
Collapse
|
3
|
Huang W, Zhang Y, Zhou R. Induced pluripotent stem cell for modeling Pompe disease. Front Cardiovasc Med 2022; 9:1061384. [PMID: 36620633 PMCID: PMC9815144 DOI: 10.3389/fcvm.2022.1061384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Pompe disease (PD) is a rare, autosomal recessive, inherited, and progressive metabolic disorder caused by α-glucosidase defect in lysosomes, resulting in abnormal glycogen accumulation. Patients with PD characteristically have multisystem pathological disorders, particularly hypertrophic cardiomyopathy, muscle weakness, and hepatomegaly. Although the pathogenesis and clinical outcomes of PD are well-established, disease-modeling ability, mechanism elucidation, and drug development targeting PD have been substantially limited by the unavailable PD-relevant cell models. This obstacle has been overcome with the help of induced pluripotent stem cell (iPSC) reprogramming technology, thus providing a powerful tool for cell replacement therapy, disease modeling, drug screening, and drug toxicity assessment. This review focused on the exciting achievement of PD disease modeling and mechanism exploration using iPSC.
Collapse
Affiliation(s)
- Wenjun Huang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanmin Zhang
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China,Department of Cardiology, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Zhou
- National Regional Children's Medical Center (Northwest), Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, China,*Correspondence: Rui Zhou ✉
| |
Collapse
|
4
|
Gonzalez TJ, Simon KE, Blondel LO, Fanous MM, Roger AL, Maysonet MS, Devlin GW, Smith TJ, Oh DK, Havlik LP, Castellanos Rivera RM, Piedrahita JA, ElMallah MK, Gersbach CA, Asokan A. Cross-species evolution of a highly potent AAV variant for therapeutic gene transfer and genome editing. Nat Commun 2022; 13:5947. [PMID: 36210364 PMCID: PMC9548504 DOI: 10.1038/s41467-022-33745-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022] Open
Abstract
Recombinant adeno-associated viral (AAV) vectors are a promising gene delivery platform, but ongoing clinical trials continue to highlight a relatively narrow therapeutic window. Effective clinical translation is confounded, at least in part, by differences in AAV biology across animal species. Here, we tackle this challenge by sequentially evolving AAV capsid libraries in mice, pigs and macaques. We discover a highly potent, cross-species compatible variant (AAV.cc47) that shows improved attributes benchmarked against AAV serotype 9 as evidenced by robust reporter and therapeutic gene expression, Cre recombination and CRISPR genome editing in normal and diseased mouse models. Enhanced transduction efficiency of AAV.cc47 vectors is further corroborated in macaques and pigs, providing a strong rationale for potential clinical translation into human gene therapies. We envision that ccAAV vectors may not only improve predictive modeling in preclinical studies, but also clinical translatability by broadening the therapeutic window of AAV based gene therapies.
Collapse
Affiliation(s)
- Trevor J Gonzalez
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Katherine E Simon
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - Leo O Blondel
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Marco M Fanous
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Angela L Roger
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | | | - Garth W Devlin
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Timothy J Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Daniel K Oh
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - L Patrick Havlik
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | | | - Jorge A Piedrahita
- North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - Mai K ElMallah
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Charles A Gersbach
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Aravind Asokan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Roger AL, Sethi R, Huston ML, Scarrow E, Bao-Dai J, Lai E, Biswas DD, Haddad LE, Strickland LM, Kishnani PS, ElMallah MK. What's new and what's next for gene therapy in Pompe disease? Expert Opin Biol Ther 2022; 22:1117-1135. [PMID: 35428407 PMCID: PMC10084869 DOI: 10.1080/14712598.2022.2067476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/14/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pompe disease is an autosomal recessive disorder caused by a deficiency of acid-α-glucosidase (GAA), an enzyme responsible for hydrolyzing lysosomal glycogen. A lack of GAA leads to accumulation of glycogen in the lysosomes of cardiac, skeletal, and smooth muscle cells, as well as in the central and peripheral nervous system. Enzyme replacement therapy has been the standard of care for 15 years and slows disease progression, particularly in the heart, and improves survival. However, there are limitations of ERT success, which gene therapy can overcome. AREAS COVERED Gene therapy offers several advantages including prolonged and consistent GAA expression and correction of skeletal muscle as well as the critical CNS pathology. We provide a systematic review of the preclinical and clinical outcomes of adeno-associated viral mediated gene therapy and alternative gene therapy strategies, highlighting what has been successful. EXPERT OPINION Although the preclinical and clinical studies so far have been promising, barriers exist that need to be addressed in gene therapy for Pompe disease. New strategies including novel capsids for better targeting, optimized DNA vectors, and adjuctive therapies will allow for a lower dose, and ameliorate the immune response.
Collapse
Affiliation(s)
- Angela L. Roger
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Ronit Sethi
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Meredith L. Huston
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Evelyn Scarrow
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Joy Bao-Dai
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Elias Lai
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Debolina D. Biswas
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Léa El Haddad
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Laura M. Strickland
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, North Carolina USA
| | - Mai K. ElMallah
- Division of Pulmonary Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| |
Collapse
|
6
|
Benevides ES, Sunshine MD, Rana S, Fuller DD. Optogenetic activation of the diaphragm. Sci Rep 2022; 12:6503. [PMID: 35444167 PMCID: PMC9021282 DOI: 10.1038/s41598-022-10240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Impaired diaphragm activation is common in many neuromuscular diseases. We hypothesized that expressing photoreceptors in diaphragm myofibers would enable light stimulation to evoke functional diaphragm activity, similar to endogenous bursts. In a mouse model, adeno-associated virus (AAV) encoding channelrhodopsin-2 (AAV9-CAG-ChR2-mVenus, 6.12 × 1011 vg dose) was delivered to the diaphragm using a minimally invasive method of microinjection to the intrapleural space. At 8-18 weeks following AAV injection, mice were anesthetized and studied during spontaneous breathing. We first showed that diaphragm electromyographic (EMG) potentials could be evoked with brief presentations of light, using a 473 nm high intensity LED. Evoked potential amplitude increased with intensity or duration of the light pulse. We next showed that in a paralyzed diaphragm, trains of light pulses evoked diaphragm EMG activity which resembled endogenous bursting, and this was sufficient to generate respiratory airflow. Light-evoked diaphragm EMG bursts showed no diminution after up to one hour of stimulation. Histological evaluation confirmed transgene expression in diaphragm myofibers. We conclude that intrapleural delivery of AAV9 can drive expression of ChR2 in the diaphragm and subsequent photostimulation can evoke graded compound diaphragm EMG activity similar to endogenous inspiratory bursting.
Collapse
Affiliation(s)
- Ethan S Benevides
- Rehabilitation Science PhD Program, University of Florida, Gainesville, Florida, USA.,Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Michael D Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA.,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA. .,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA. .,McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
7
|
Chen Y, Gong Y, Dou L, Zhou X, Zhang Y. Bioinformatics analysis methods for cell-free DNA. Comput Biol Med 2022; 143:105283. [PMID: 35149459 DOI: 10.1016/j.compbiomed.2022.105283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
Abstract
As a kind of novel non-invasive marker for molecular detection, cell-free DNA (cfDNA) has potential value for the early diagnosis of diseases, prognosis assessment, and efficacy monitoring. The constant developments in molecular biology detection technologies have led to an increase in clinical studies on the use of cfDNA detection methods for patients, and many gratifying outcomes have been achieved. In this review, the contributions of bioinformatics tools to the study of cfDNA are well discussed. The focus of the review is on cfDNA identification signals, cfDNA identification methods, and the relationship of cfDNA with human diseases such as hepatic cancer, lung cancer, end-stage kidney disease, and ischemic stroke. The research significance and existing problems of using cfDNA as a biomarker for diseases are also discussed.
Collapse
Affiliation(s)
- Yaojia Chen
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Yuxin Gong
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China; School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Lijun Dou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China; School of Automotive and Transportation Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Xun Zhou
- Beidahuang Industry Group General Hospital, Harbin, China.
| | - Ying Zhang
- Department of Anesthesiology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, China.
| |
Collapse
|
8
|
Han S, Wang N, Guo Y, Tang F, Xu L, Ju Y, Shi L. Application of Sparse Representation in Bioinformatics. Front Genet 2021; 12:810875. [PMID: 34976030 PMCID: PMC8715914 DOI: 10.3389/fgene.2021.810875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 11/15/2022] Open
Abstract
Inspired by L1-norm minimization methods, such as basis pursuit, compressed sensing, and Lasso feature selection, in recent years, sparse representation shows up as a novel and potent data processing method and displays powerful superiority. Researchers have not only extended the sparse representation of a signal to image presentation, but also applied the sparsity of vectors to that of matrices. Moreover, sparse representation has been applied to pattern recognition with good results. Because of its multiple advantages, such as insensitivity to noise, strong robustness, less sensitivity to selected features, and no “overfitting” phenomenon, the application of sparse representation in bioinformatics should be studied further. This article reviews the development of sparse representation, and explains its applications in bioinformatics, namely the use of low-rank representation matrices to identify and study cancer molecules, low-rank sparse representations to analyze and process gene expression profiles, and an introduction to related cancers and gene expression profile database.
Collapse
Affiliation(s)
- Shuguang Han
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Ning Wang
- Beidahuang Industry Group General Hospital, Harbin, China
| | - Yuxin Guo
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
- School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Furong Tang
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Ying Ju
- School of Informatics, Xiamen University, Xiamen, China
- *Correspondence: Ying Ju, ; Lei Shi,
| | - Lei Shi
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Ying Ju, ; Lei Shi,
| |
Collapse
|
9
|
Zulfiqar H, Sun ZJ, Huang QL, Yuan SS, Lv H, Dao FY, Lin H, Li YW. Deep-4mCW2V: A sequence-based predictor to identify N4-methylcytosine sites in Escherichia coli. Methods 2021; 203:558-563. [PMID: 34352373 DOI: 10.1016/j.ymeth.2021.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022] Open
Abstract
N4-methylcytosine (4mC) is a type of DNA modification which could regulate several biological progressions such as transcription regulation, replication and gene expressions. Precisely recognizing 4mC sites in genomic sequences can provide specific knowledge about their genetic roles. This study aimed to develop a deep learning-based model to predict 4mC sites in the Escherichia coli. In the model, DNA sequences were encoded by word embedding technique 'word2vec'. The obtained features were inputted into 1-D convolutional neural network (CNN) to discriminate 4mC sites from non-4mC sites in Escherichia coli genome. The examination on independent dataset showed that our model could yield the overall accuracy of 0.861, which was about 4.3% higher than the existing model. To provide convenience to scholars, we provided the data and source code of the model which can be freely download from https://github.com/linDing-groups/Deep-4mCW2V.
Collapse
Affiliation(s)
- Hasan Zulfiqar
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zi-Jie Sun
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Qin-Lai Huang
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Shi-Shi Yuan
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hao Lv
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Fu-Ying Dao
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hao Lin
- Center for Informational Biology and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Yan-Wen Li
- School of Information Science and Technology, Northeast Normal University, Changchun 130117, China; Key Laboratory of Intelligent Information Processing of Jilin Province, Northeast Normal University, Changchun 130117, China; Institute of Computational Biology, Northeast Normal University, Changchun 130117, China.
| |
Collapse
|
10
|
Phenotypic implications of pathogenic variant types in Pompe disease. J Hum Genet 2021; 66:1089-1099. [PMID: 33972680 DOI: 10.1038/s10038-021-00935-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/01/2021] [Accepted: 04/20/2021] [Indexed: 11/08/2022]
Abstract
Newborn screening and therapies for Pompe disease (glycogen storage disease type II, acid maltase deficiency) will continue to expand in the future. It is thus important to determine whether enzyme activity or type of pathogenic genetic variant in GAA can best predict phenotypic severity, particularly the presence of infantile-onset Pompe disease (IOPD) versus late-onset Pompe disease (LOPD). We performed a retrospective analysis of 23 participants with genetically-confirmed cases of Pompe disease. The following data were collected: clinical details including presence or absence of cardiomyopathy, enzyme activity levels, and features of GAA variants including exon versus intron location and splice site versus non-splice site. Several combinations of GAA variant types for individual participants had significant associations with disease subtype, cardiomyopathy, age at diagnosis, gross motor function scale (GMFS), and stability of body weight. The presence of at least one splice site variant (c.546 G > C/p.T182 = , c.1076-22 T > G, c.2646 + 2 T > A, and the classic c.-32-13T > G variant) was associated with LOPD, while the presence of non-splice site variants on both alleles was associated with IOPD. Enzyme activity levels in isolation were not sufficient to predict disease subtype or other major clinical features. To extend the findings of prior studies, we found that multiple types of splice site variants beyond the classic c.-32-13T > G variant are often associated with a milder phenotype. Enzyme activity levels continue to have utility for supporting the diagnosis when the genetic variants are ambiguous. It is important for newly diagnosed patients with Pompe disease to have complete genetic, cardiac, and neurological evaluations.
Collapse
|
11
|
McCall AL, Dhindsa JS, Bailey AM, Pucci LA, Strickland LM, ElMallah MK. Glycogen accumulation in smooth muscle of a Pompe disease mouse model. J Smooth Muscle Res 2021; 57:8-18. [PMID: 33883348 PMCID: PMC8053439 DOI: 10.1540/jsmr.57.8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Pompe disease is a lysosomal storage disease caused by mutations within the
GAA gene, which encodes acid α-glucosidase (GAA)—an enzyme necessary
for lysosomal glycogen degradation. A lack of GAA results in an accumulation of glycogen
in cardiac and skeletal muscle, as well as in motor neurons. The only FDA approved
treatment for Pompe disease—an enzyme replacement therapy (ERT)—increases survival of
patients, but has unmasked previously unrecognized clinical manifestations of Pompe
disease. These clinical signs and symptoms include tracheo-bronchomalacia, vascular
aneurysms, and gastro-intestinal discomfort. Together, these previously unrecognized
pathologies indicate that GAA-deficiency impacts smooth muscle in addition to skeletal and
cardiac muscle. Thus, we sought to characterize smooth muscle pathology in the airway,
vascular, gastrointestinal, and genitourinary in the Gaa−/−
mouse model. Increased levels of glycogen were present in smooth muscle cells of the
aorta, trachea, esophagus, stomach, and bladder of Gaa−/−
mice, compared to wild type mice. In addition, there was an increased
abundance of both lysosome membrane protein (LAMP1) and autophagosome membrane protein
(LC3) indicating vacuolar accumulation in several tissues. Taken together, we show that
GAA deficiency results in subsequent pathology in smooth muscle cells, which may lead to
life-threatening complications if not properly treated.
Collapse
Affiliation(s)
- Angela L McCall
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Justin S Dhindsa
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Aidan M Bailey
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Logan A Pucci
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Laura M Strickland
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Mai K ElMallah
- Division of Pulmonary Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
12
|
Salabarria SM, Nair J, Clement N, Smith BK, Raben N, Fuller DD, Byrne BJ, Corti M. Advancements in AAV-mediated Gene Therapy for Pompe Disease. J Neuromuscul Dis 2020; 7:15-31. [PMID: 31796685 PMCID: PMC7029369 DOI: 10.3233/jnd-190426] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pompe disease (glycogen storage disease type II) is caused by mutations in acid α-glucosidase (GAA) resulting in lysosomal pathology and impairment of the muscular and cardio-pulmonary systems. Enzyme replacement therapy (ERT), the only approved therapy for Pompe disease, improves muscle function by reducing glycogen accumulation but this approach entails several limitations including a short drug half-life and an antibody response that results in reduced efficacy. To address these limitations, new treatments such as gene therapy are under development to increase the intrinsic ability of the affected cells to produce GAA. Key components to gene therapy strategies include the choice of vector, promoter, and the route of administration. The efficacy of gene therapy depends on the ability of the vector to drive gene expression in the target tissue and also on the recipient's immune tolerance to the transgene protein. In this review, we discuss the preclinical and clinical studies that are paving the way for the development of a gene therapy strategy for patients with early and late onset Pompe disease as well as some of the challenges for advancing gene therapy.
Collapse
Affiliation(s)
- S M Salabarria
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - J Nair
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - N Clement
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - B K Smith
- Department of Physical Therapy and Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | - N Raben
- Laboratory of Protein Trafficking and Organelle Biology, Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - D D Fuller
- Department of Physical Therapy and Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida, USA
| | - B J Byrne
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| | - M Corti
- Department of Pediatrics and Powell Gene Therapy Center, University of Florida, Gainesville, Floria, USA
| |
Collapse
|
13
|
Han SO, Li S, McCall A, Arnson B, Everitt JI, Zhang H, Young SP, ElMallah MK, Koeberl DD. Comparisons of Infant and Adult Mice Reveal Age Effects for Liver Depot Gene Therapy in Pompe Disease. Mol Ther Methods Clin Dev 2020; 17:133-142. [PMID: 31909086 PMCID: PMC6938806 DOI: 10.1016/j.omtm.2019.11.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/26/2019] [Indexed: 01/20/2023]
Abstract
Pompe disease is caused by the deficiency of lysosomal acid α-glucosidase (GAA). It is expected that gene therapy to replace GAA with adeno-associated virus (AAV) vectors will be less effective early in life because of the rapid loss of vector genomes. AAV2/8-LSPhGAA (3 × 1010 vector genomes [vg]/mouse) was administered to infant (2-week-old) or adult (2-month-old) GAA knockout mice. AAV vector transduction in adult mice significantly corrected GAA deficiency in the heart (p < 0.0001), diaphragm (p < 0.01), and quadriceps (p < 0.001) for >50 weeks. However, in infant mice, the same treatment only partially corrected GAA deficiency in the heart (p < 0.05), diaphragm (p < 0.05), and quadriceps (p < 0.05). The clearance of glycogen was much more efficient in adult mice compared with infant mice. Improved wire hang test latency was observed for treated adults (p < 0.05), but not for infant mice. Abnormal ventilation was corrected in both infant and adult mice. Vector-treated female mice demonstrated functional improvement, despite a lower degree of biochemical correction compared with male mice. The relative vector dose for infants was approximately 3-fold higher than adults, when normalized to body weight at the time of vector administration. Given these data, the dose requirement to achieve similar efficacy will be higher for the treatment of young patients.
Collapse
Affiliation(s)
- Sang-oh Han
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Songtao Li
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Angela McCall
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Benjamin Arnson
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey I. Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Haoyue Zhang
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Sarah P. Young
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Mai K. ElMallah
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dwight D. Koeberl
- Division of Medical Genetics, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Molecular Genetics and Metabolism, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
14
|
Cagin U, Puzzo F, Gomez MJ, Moya-Nilges M, Sellier P, Abad C, Van Wittenberghe L, Daniele N, Guerchet N, Gjata B, Collaud F, Charles S, Sola MS, Boyer O, Krijnse-Locker J, Ronzitti G, Colella P, Mingozzi F. Rescue of Advanced Pompe Disease in Mice with Hepatic Expression of Secretable Acid α-Glucosidase. Mol Ther 2020; 28:2056-2072. [PMID: 32526204 PMCID: PMC7474269 DOI: 10.1016/j.ymthe.2020.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/15/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Pompe disease is a neuromuscular disorder caused by disease-associated variants in the gene encoding for the lysosomal enzyme acid α-glucosidase (GAA), which converts lysosomal glycogen to glucose. We previously reported full rescue of Pompe disease in symptomatic 4-month-old Gaa knockout (Gaa−/−) mice by adeno-associated virus (AAV) vector-mediated liver gene transfer of an engineered secretable form of GAA (secGAA). Here, we showed that hepatic expression of secGAA rescues the phenotype of 4-month-old Gaa−/− mice at vector doses at which the native form of GAA has little to no therapeutic effect. Based on these results, we then treated severely affected 9-month-old Gaa−/− mice with an AAV vector expressing secGAA and followed the animals for 9 months thereafter. AAV-treated Gaa−/− mice showed complete reversal of the Pompe phenotype, with rescue of glycogen accumulation in most tissues, including the central nervous system, and normalization of muscle strength. Transcriptomic profiling of skeletal muscle showed rescue of most altered pathways, including those involved in mitochondrial defects, a finding supported by structural and biochemical analyses, which also showed restoration of lysosomal function. Together, these results provide insight into the reversibility of advanced Pompe disease in the Gaa−/− mouse model via liver gene transfer of secGAA.
Collapse
Affiliation(s)
- Umut Cagin
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Francesco Puzzo
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France
| | - Manuel Jose Gomez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Pauline Sellier
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Catalina Abad
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Nathalie Daniele
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Nicolas Guerchet
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Bernard Gjata
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Severine Charles
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Marcelo Simon Sola
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Olivier Boyer
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Pasqualina Colella
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Federico Mingozzi
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France; Spark Therapeutics, Philadelphia, PA 19103, USA.
| |
Collapse
|
15
|
Fusco AF, McCall AL, Dhindsa JS, Zheng L, Bailey A, Kahn AF, ElMallah MK. The Respiratory Phenotype of Pompe Disease Mouse Models. Int J Mol Sci 2020; 21:ijms21062256. [PMID: 32214050 PMCID: PMC7139647 DOI: 10.3390/ijms21062256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 01/10/2023] Open
Abstract
Pompe disease is a glycogen storage disease caused by a deficiency in acid α-glucosidase (GAA), a hydrolase necessary for the degradation of lysosomal glycogen. This deficiency in GAA results in muscle and neuronal glycogen accumulation, which causes respiratory insufficiency. Pompe disease mouse models provide a means of assessing respiratory pathology and are important for pre-clinical studies of novel therapies that aim to treat respiratory dysfunction and improve quality of life. This review aims to compile and summarize existing manuscripts that characterize the respiratory phenotype of Pompe mouse models. Manuscripts included in this review were selected utilizing specific search terms and exclusion criteria. Analysis of these findings demonstrate that Pompe disease mouse models have respiratory physiological defects as well as pathologies in the diaphragm, tongue, higher-order respiratory control centers, phrenic and hypoglossal motor nuclei, phrenic and hypoglossal nerves, neuromuscular junctions, and airway smooth muscle. Overall, the culmination of these pathologies contributes to severe respiratory dysfunction, underscoring the importance of characterizing the respiratory phenotype while developing effective therapies for patients.
Collapse
|
16
|
Bragato C, Carra S, Blasevich F, Salerno F, Brix A, Bassi A, Beltrame M, Cotelli F, Maggi L, Mantegazza R, Mora M. Glycogen storage in a zebrafish Pompe disease model is reduced by 3-BrPA treatment. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165662. [PMID: 31917327 DOI: 10.1016/j.bbadis.2020.165662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/16/2019] [Accepted: 01/02/2020] [Indexed: 12/18/2022]
Abstract
Pompe disease (PD) is an autosomal recessive muscular disorder caused by deficiency of the glycogen hydrolytic enzyme acid α-glucosidase (GAA). The enzyme replacement therapy, currently the only available therapy for PD patients, is efficacious in improving cardiomyopathy in the infantile form, but not equally effective in the late onset cases with involvement of skeletal muscle. Correction of the skeletal muscle phenotype has indeed been challenging, probably due to concomitant dysfunctional autophagy. The increasing attention to the pathogenic mechanisms of PD and the search of new therapeutic strategies prompted us to generate and characterize a novel transient PD model, using zebrafish. Our model presented increased glycogen content, markedly altered motor behavior and increased lysosome content, in addition to altered expression of the autophagy-related transcripts and proteins Beclin1, p62 and Lc3b. Furthermore, the model was used to assess the beneficial effects of 3-bromopyruvic acid (3-BrPA). Treatment with 3-BrPA induced amelioration of the model phenotypes regarding glycogen storage, motility behavior and autophagy-related transcripts and proteins. Our zebrafish PD model recapitulates most of the defects observed in human patients, proving to be a powerful translational model. Moreover, 3-BrPA unveiled to be a promising compound for treatment of conditions with glycogen accumulation.
Collapse
Affiliation(s)
- Cinzia Bragato
- PhD program in Neuroscience, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy; Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy.
| | - Silvia Carra
- Laboratory of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, Milan, 20149, Italy
| | - Flavia Blasevich
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Franco Salerno
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Alessia Brix
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Andrea Bassi
- Department of Physics, Politecnico di Milano, Piazza Leonardo da Vinci 32, Milan, 20133, Italy
| | - Monica Beltrame
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, Milan, 20133, Italy
| | - Franco Cotelli
- Department of Biosciences, Università degli Studi di Milano, Via Celoria 26, Milan, 20133, Italy
| | - Lorenzo Maggi
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Renato Mantegazza
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy.
| |
Collapse
|