1
|
Jiao D, Xu L, Gu Z, Yan H, Shen D, Gu X. Pathogenesis, diagnosis, and treatment of epilepsy: electromagnetic stimulation-mediated neuromodulation therapy and new technologies. Neural Regen Res 2025; 20:917-935. [PMID: 38989927 PMCID: PMC11438347 DOI: 10.4103/nrr.nrr-d-23-01444] [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: 08/28/2023] [Revised: 10/31/2023] [Accepted: 01/18/2024] [Indexed: 07/12/2024] Open
Abstract
Epilepsy is a severe, relapsing, and multifactorial neurological disorder. Studies regarding the accurate diagnosis, prognosis, and in-depth pathogenesis are crucial for the precise and effective treatment of epilepsy. The pathogenesis of epilepsy is complex and involves alterations in variables such as gene expression, protein expression, ion channel activity, energy metabolites, and gut microbiota composition. Satisfactory results are lacking for conventional treatments for epilepsy. Surgical resection of lesions, drug therapy, and non-drug interventions are mainly used in clinical practice to treat pain associated with epilepsy. Non-pharmacological treatments, such as a ketogenic diet, gene therapy for nerve regeneration, and neural regulation, are currently areas of research focus. This review provides a comprehensive overview of the pathogenesis, diagnostic methods, and treatments of epilepsy. It also elaborates on the theoretical basis, treatment modes, and effects of invasive nerve stimulation in neurotherapy, including percutaneous vagus nerve stimulation, deep brain electrical stimulation, repetitive nerve electrical stimulation, in addition to non-invasive transcranial magnetic stimulation and transcranial direct current stimulation. Numerous studies have shown that electromagnetic stimulation-mediated neuromodulation therapy can markedly improve neurological function and reduce the frequency of epileptic seizures. Additionally, many new technologies for the diagnosis and treatment of epilepsy are being explored. However, current research is mainly focused on analyzing patients' clinical manifestations and exploring relevant diagnostic and treatment methods to study the pathogenesis at a molecular level, which has led to a lack of consensus regarding the mechanisms related to the disease.
Collapse
Affiliation(s)
- Dian Jiao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Lai Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hua Yan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Dingding Shen
- Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| |
Collapse
|
2
|
Jehi L. Advances in Therapy for Refractory Epilepsy. Annu Rev Med 2025; 76:389-402. [PMID: 39532109 DOI: 10.1146/annurev-med-050522-034458] [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: 11/16/2024]
Abstract
Drug-resistant epilepsy (DRE) is defined as failure to achieve sustained seizure control with adequate trials of two appropriate antiseizure medications (ASMs). DRE affects one-third of patients with epilepsy and is associated with significant morbidity and mortality. Newer ASMs provide pharmacological therapy that is better tolerated but not necessarily more effective than older ASMs. Resective brain surgery is the gold standard to treat DRE and achieve seizure freedom, with laser ablation offering an alternative with less morbidity but lower effectiveness. For patients who are not candidates for resection or ablation, multiple neuromodulation options can reduce seizure burden. These neuromodulation devices have shown comparable effectiveness in randomized clinical trials, but the results vary in open-label follow-up cohorts, as do the risks of complications and associated costs. Dietary therapies can help, particularly in pediatric genetic epilepsies. Innovative genetic therapy approaches are being pursued, offering the promise of precision medicine.
Collapse
Affiliation(s)
- Lara Jehi
- Epilepsy Center, Cleveland Clinic, Cleveland, Ohio, USA;
| |
Collapse
|
3
|
González Molina LA, Dolga AM, Rots MG, Sarno F. The Promise of Epigenetic Editing for Treating Brain Disorders. Subcell Biochem 2025; 108:111-190. [PMID: 39820862 DOI: 10.1007/978-3-031-75980-2_4] [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: 01/19/2025]
Abstract
Brain disorders, especially neurodegenerative diseases, affect millions of people worldwide. There is no causal treatment available; therefore, there is an unmet clinical need for finding therapeutic options for these diseases. Epigenetic research has resulted in identification of various genomic loci with differential disease-specific epigenetic modifications, mainly DNA methylation. These biomarkers, although not yet translated into clinically approved options, offer therapeutic targets as epigenetic modifications are reversible. Indeed, clinical trials are designed to inhibit epigenetic writers, erasers, or readers using epigenetic drugs to interfere with epigenetic dysregulation in brain disorders. However, since such drugs elicit genome-wide effects and potentially cause toxicity, the recent developments in the field of epigenetic editing are gaining widespread attention. In this review, we provide examples of epigenetic biomarkers and epi-drugs, while describing efforts in the field of epigenetic editing, to eventually make a difference for the currently incurable brain disorders.
Collapse
Affiliation(s)
- Luis A González Molina
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Marianne G Rots
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Federica Sarno
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Samanta D. A comprehensive review of evolving treatment strategies for Dravet syndrome: Insights from randomized trials, meta-analyses, real-world evidence, and emerging therapeutic approaches. Epilepsy Behav 2025; 162:110171. [PMID: 39612634 DOI: 10.1016/j.yebeh.2024.110171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
Dravet syndrome (DS) is a severe genetic developmental and epileptic encephalopathy, primarily caused by SCN1A gene mutations. Historically, treatments like clobazam and valproate have been used without evidence from randomized controlled trials (RCTs). However, the therapeutic landscape of DS has evolved with multiple RCTs demonstrating the efficacy and safety of three antiseizure medications (ASMs): stiripentol, cannabidiol (CBD), and fenfluramine. In the absence of direct comparisons between these therapies, several network meta-analyses have been conducted to compare the ASMs, while expert consensus has independently been developed to formulate treatment guidelines. While these three ASMs show promise in reducing seizures, increasing awareness of non-seizure outcomes-such as cognitive development and quality of life-has shifted the focus of evaluation. Some recent real-world studies of these ASMs have reported improvements in these non-seizure outcomes, alongside sustained efficacy and safety. However, natural history studies continue to underscore persistent deficits in these areas and highlight suboptimal long-term seizure control despite the use of these therapies. This review addresses these gaps by first discussing network meta-analyses and treatment guidelines, along with the practical limitations of these approaches. It then examines the long-term efficacy, safety, non-seizure effects, and cost-effectiveness from real-world studies of these ASMs. Finally, emerging research on novel therapeutic approaches, including genetic and serotonergic modulation, is explored. By evaluating these developments, this review aims to guide clinical decision-making and propose future directions for optimizing DS care.
Collapse
Affiliation(s)
- Debopam Samanta
- Division of Child Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
5
|
Zhou L, Wang Y, Xu Y, Zhang Y, Zhu C. Advances in AAV-mediated gene replacement therapy for pediatric monogenic neurological disorders. Mol Ther Methods Clin Dev 2024; 32:101357. [PMID: 39559557 PMCID: PMC11570947 DOI: 10.1016/j.omtm.2024.101357] [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] [Indexed: 11/20/2024]
Abstract
Pediatric monogenetic diseases encompass a spectrum of debilitating neurological disorders that affect infants and children, often resulting in profound cognitive and motor impairments. Gene replacement therapy holds immense promise in addressing the underlying genetic defects responsible for these conditions. Adeno-associated virus (AAV) vectors have emerged as a leading platform for delivering therapeutic genes due to their safety profile and ability to transduce various cell types, including neurons. This review highlights recent advancements in AAV-mediated gene replacement therapy for pediatric monogenetic diseases, focusing on key preclinical and clinical studies. We discuss various strategies to enhance transduction efficiency, target specificity, and safety. Furthermore, we explore challenges such as immune responses, along with innovative approaches to overcome these obstacles. Moreover, we examine the clinical outcomes and safety profiles of AAV-based gene therapies in pediatric patients, providing insights into the feasibility and efficacy of these interventions. Finally, we discuss future directions and potential avenues for further research to optimize the therapeutic potential of AAV-delivered gene replacement therapy for pediatric encephalopathies, ultimately aiming to improve the quality of life for affected individuals and their families.
Collapse
Affiliation(s)
- Livia Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
6
|
Sang Y, Xu L, Bao Z. Development of artificial transcription factors and their applications in cell reprograming, genetic screen, and disease treatment. Mol Ther 2024; 32:4208-4234. [PMID: 39473180 PMCID: PMC11638881 DOI: 10.1016/j.ymthe.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/18/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024] Open
Abstract
Gene dysregulations are associated with many human diseases, such as cancers and hereditary diseases. Artificial transcription factors (ATFs) are synthetic molecular tools to regulate the expression of disease-associated genes, which is of great significance in basic biological research and biomedical applications. Recent advances in the engineering of ATFs for regulating endogenous gene expression provide an expanded set of tools for understanding and treating diseases. However, the potential immunogenicity, large size, inefficient delivery, and off-target effects persist as obstacles for ATFs to be developed into therapeutics. Moreover, the activation of an endogenous gene following ATF activity lacks durability. In this review, we first describe the functional components of ATFs, including DNA-binding domains, transcriptional effector domains, and control switches. We then highlight examples of applications of ATFs, including cell reprogramming and differentiation, pathogenic gene screening, and disease treatment. Finally, we analyze and summarize major challenges for the clinical translation of ATFs and propose potential strategies to improve these useful molecular tools.
Collapse
Affiliation(s)
- Yetong Sang
- Institute of Bioengineering & Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, Zhejiang, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, Zhejiang, China
| | - Lingjie Xu
- Institute of Bioengineering & Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, Zhejiang, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, Zhejiang, China
| | - Zehua Bao
- Institute of Bioengineering & Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, Zhejiang, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, Zhejiang, China; Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
7
|
Griflyuk AV, Postnikova TY, Zaitsev AV. Animal Models of Febrile Seizures: Limitations and Recent Advances in the Field. Cells 2024; 13:1895. [PMID: 39594643 PMCID: PMC11592604 DOI: 10.3390/cells13221895] [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: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Febrile seizures (FSs) are defined as seizures occurring in children aged 6 months to 5 years with a background of elevated body temperature. It is one of the most common neurological disorders of childhood, emphasizing the importance of understanding the causes of FSs and their impact on the developing nervous system. However, there are significant limitations to the technologies currently available for studying the etiology and pathophysiology of seizures in humans. It is currently not possible to adequately capture the subtle molecular and structural rearrangements of the nervous system that can occur after seizures in humans. The use of animal models can be invaluable for these purposes. The most commonly used models in modern research are hyperthermic models in rats and mice aged 10-12 days. While these models can reproduce many of the characteristics of FSs, they have certain limitations. This review outlines the key considerations when working with models of FSs, provides an overview of current approaches to producing seizures in different model subjects, and presents a summary of key findings regarding morphological and functional changes in the brain and behavioral alterations that have been identified in studies using animal models of FSs.
Collapse
Affiliation(s)
| | | | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 44, Toreza Prospekt, Saint Petersburg 194223, Russia; (A.V.G.); (T.Y.P.)
| |
Collapse
|
8
|
Specchio N, Trivisano M, Aronica E, Balestrini S, Arzimanoglou A, Colasante G, Cross JH, Jozwiak S, Wilmshurst JM, Vigevano F, Auvin S, Nabbout R, Curatolo P. The expanding field of genetic developmental and epileptic encephalopathies: current understanding and future perspectives. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:821-834. [PMID: 39419567 DOI: 10.1016/s2352-4642(24)00196-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 10/19/2024]
Abstract
Recent advances in genetic testing technologies have revolutionised the identification of genetic abnormalities in early onset developmental and epileptic encephalopathies (DEEs). In this Review, we provide an update on the expanding landscape of genetic factors contributing to DEEs, encompassing over 800 reported genes. We focus on the cellular and molecular mechanisms driving epileptogenesis, with an emphasis on emerging therapeutic strategies and effective treatment options. We explore noteworthy, novel genes linked to DEE phenotypes, such as gBRAT-1 and GNAO1, and gene families such as GRIN and HCN. Understanding the network-level effects of gene variants will pave the way for potential gene therapy applications. Given the diverse comorbidities associated with DEEs, a multidisciplinary team approach is essential. Despite ongoing efforts and improved genetic testing, DEEs lack a cure, and treatment complexities persist. This Review underscores the necessity for larger international prospective studies focusing on both seizure outcomes and developmental trajectories.
Collapse
Affiliation(s)
- Nicola Specchio
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesu' Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies, EpiCARE, Rome, Italy.
| | - Marina Trivisano
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesu' Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies, EpiCARE, Rome, Italy
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Department of (Neuro)Pathology, Amsterdam, Netherlands; Stichting Epilepsie Instellingen Nederland, Heemstede, Netherlands
| | - Simona Balestrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies, EpiCARE, Florence, Italy; Neuroscience Department, University of Florence, Florence, Italy; Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Alexis Arzimanoglou
- Paediatric Epilepsy and Neurophysiology Department, Hospital San Juan de Dios, Coordinating member of the European Reference Network on Rare and Complex Epilepsies, EpiCARE, Barcelona, Spain
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - J Helen Cross
- UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
| | - Sergiusz Jozwiak
- Research Department, Children's Memorial Health Institute, EpicARE Member, Warsaw, Poland
| | - Jo M Wilmshurst
- Department of Paediatric Neurology, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, South Africa
| | - Federico Vigevano
- Pediatric Neurorehabilitation Department, IRCCS San Raffaele, Rome, Italy
| | - Stéphane Auvin
- AP-HP, Service de Neurologie Pédiatrique, Centre de référence Epilepsies Rares, Member of European Reference Network EpiCARE, Hôpital Universitaire Robert-Debré, Paris, France; Université Paris-Cité, INSERM Neuro Diderot, Paris, France; Institut Universitaire de France, Paris, France
| | - Rima Nabbout
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker-Enfants Malades Hospital, AP-HP, Member of European Reference Network EpiCARE, Institut Imagine, INSERM, UMR 1163, Université de Paris Cité, Paris, France
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| |
Collapse
|
9
|
Yang Z, Li H, Luo M, Yi H, Han X, Liu E, Yao S, Hu Z. Identification of c.146G > A mutation in a Fabry patient and its correction by customized Cas9 base editors in vitro. Int J Biol Macromol 2024; 282:136922. [PMID: 39490876 DOI: 10.1016/j.ijbiomac.2024.136922] [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: 09/05/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Fabry disease (FD) is a rare X-linked lysosomal storage disorder caused by mutations in the GLA gene, leading to reduced α-galactosidase (α-Gal A) activity. Current treatments, like enzyme replacement, have limitations affecting efficacy and patient outcomes. CRISPR/Cas9 genome editing tools may offer the potential to develop therapeutic strategy via correcting GLA mutations. In this study, we diagnosed a female FD patient with a missense mutation in exon 1 of the GLA gene (c.146G > A, p.R49H). Bioinformatic predictions and biochemical analyses in GLA-knockout cells revealed that this mutation significantly reduced α-Gal A stability and activity, confirming its pathogenicity. To correct this, we used adenine base editing. The mutation, along with a nearby bystander A, was efficiently edited by the traditional N-terminal adenine base editor. To avoid unwanted bystander editing, we developed a series of domain-inlaid base editors with the aim of narrowing editing window. The most effective variant, with deaminase inserted between the 947th and 948th residues of the RUVC3 domain, was further optimized by modifying linker rigidity. These adjustments shifted the editing window, eliminating bystander editing. Our findings clarify the pathogenic nature of a novel GLA mutation and demonstrate the potential of a customized base editor for therapeutic application in FD.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mei Luo
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan university, Chengdu 610041, Sichuan, China
| | - Haonan Yi
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan university, Chengdu 610041, Sichuan, China
| | - Xinyu Han
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan university, Chengdu 610041, Sichuan, China
| | - Enze Liu
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan university, Chengdu 610041, Sichuan, China
| | - Shaohua Yao
- Laboratory of Biotherapy, National Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan university, Chengdu 610041, Sichuan, China.
| | - Zhangxue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
10
|
Ng BW, Kaukonen MK, McClements ME, Shamsnajafabadi H, MacLaren RE, Cehajic-Kapetanovic J. Genetic therapies and potential therapeutic applications of CRISPR activators in the eye. Prog Retin Eye Res 2024; 102:101289. [PMID: 39127142 DOI: 10.1016/j.preteyeres.2024.101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Conventional gene therapy involving supplementation only treats loss-of-function diseases and is limited by viral packaging sizes, precluding therapy of large genes. The discovery of CRISPR/Cas has led to a paradigm shift in the field of genetic therapy, with the promise of precise gene editing, thus broadening the range of diseases that can be treated. The initial uses of CRISPR/Cas have focused mainly on gene editing or silencing of abnormal variants via utilising Cas endonuclease to trigger the target cell endogenous non-homologous end joining. Subsequently, the technology has evolved to modify the Cas enzyme and even its guide RNA, leading to more efficient editing tools in the form of base and prime editing. Further advancements of this CRISPR/Cas technology itself have expanded its functional repertoire from targeted editing to programmable transactivation, shifting the therapeutic focus to precise endogenous gene activation or upregulation with the potential for epigenetic modifications. In vivo experiments using this platform have demonstrated the potential of CRISPR-activators (CRISPRa) to treat various loss-of-function diseases, as well as in regenerative medicine, highlighting their versatility to overcome limitations associated with conventional strategies. This review summarises the molecular mechanisms of CRISPRa platforms, the current applications of this technology in vivo, and discusses potential solutions to translational hurdles for this therapy, with a focus on ophthalmic diseases.
Collapse
Affiliation(s)
- Benjamin Wj Ng
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Maria K Kaukonen
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Michelle E McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Hoda Shamsnajafabadi
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Robert E MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jasmina Cehajic-Kapetanovic
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
11
|
Shang J, Song F, Zhang Z, Chen D, Yang S. Application of novel CRISPR tools in brain therapy. Life Sci 2024; 352:122855. [PMID: 38908787 DOI: 10.1016/j.lfs.2024.122855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
In recent years, the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based genome editing toolkit has been widely used to modify the genome sequence of organisms. As the CRISPR toolbox continues to grow and new CRISPR-associated (Cas) proteins are discovered, its applications have expanded beyond conventional genome editing. This now encompass epigenetic editing, gene expression control, and various other functions. Notably, these advancements are finding practical application in the treatment of brain diseases. Furthermore, the amalgamation of CRISPR and Chimeric Antigen Receptor T-cell (CAR-T) technologies has emerged as a potential approach for disease treatment. With this in mind, this review commences by offering a comprehensive overview of recent advancements in CRISPR gene editing tools. This encompasses an exploration of various Cas proteins, gene expression control, epigenetic editing, base editing and primer editing. Additionally, we present an in-depth examination of the manifold applications of these innovative CRISPR tools in the realms of brain therapeutics, such as neurodegenerative diseases, neurological syndromes and genetic disorders, epileptic disorders, and brain tumors, also explore the pathogenesis of these diseases. This includes their utilization in modeling, gene screening, therapeutic gene editing, as well as their emerging synergy with CAR-T technology. Finally, we discuss the remaining technical challenges that need to be addressed for effective utilization of CRISPR tools in disease treatment.
Collapse
Affiliation(s)
- Jiawen Shang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Fei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Di Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Sen Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| |
Collapse
|
12
|
Roth GV, Gengaro IR, Qi LS. Precision epigenetic editing: Technological advances, enduring challenges, and therapeutic applications. Cell Chem Biol 2024; 31:S2451-9456(24)00309-X. [PMID: 39137782 DOI: 10.1016/j.chembiol.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/31/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024]
Abstract
The epigenome is a complex framework through which gene expression is precisely and flexibly modulated to incorporate heritable memory and responses to environmental stimuli. It governs diverse cellular processes, including cell fate, disease, and aging. The need to understand this system and precisely control gene expression outputs for therapeutic purposes has precipitated the development of a diverse set of epigenetic editing tools. Here, we review the existing toolbox for targeted epigenetic editing, technical considerations of the current technologies, and opportunities for future development. We describe applications of therapeutic epigenetic editing and their potential for treating disease, with a discussion of ongoing delivery challenges that impede certain clinical interventions, particularly in the brain. With simultaneous advancements in available engineering tools and appropriate delivery technologies, we predict that epigenetic editing will increasingly cement itself as a powerful approach for safely treating a wide range of disorders in all tissues of the body.
Collapse
Affiliation(s)
- Goldie V Roth
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Isabella R Gengaro
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Lei S Qi
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
13
|
McCutcheon SR, Rohm D, Iglesias N, Gersbach CA. Epigenome editing technologies for discovery and medicine. Nat Biotechnol 2024; 42:1199-1217. [PMID: 39075148 DOI: 10.1038/s41587-024-02320-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/19/2024] [Indexed: 07/31/2024]
Abstract
Epigenome editing has rapidly evolved in recent years, with diverse applications that include elucidating gene regulation mechanisms, annotating coding and noncoding genome functions and programming cell state and lineage specification. Importantly, given the ubiquitous role of epigenetics in complex phenotypes, epigenome editing has unique potential to impact a broad spectrum of diseases. By leveraging powerful DNA-targeting technologies, such as CRISPR, epigenome editing exploits the heritable and reversible mechanisms of epigenetics to alter gene expression without introducing DNA breaks, inducing DNA damage or relying on DNA repair pathways.
Collapse
Affiliation(s)
- Sean R McCutcheon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Dahlia Rohm
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Nahid Iglesias
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA.
| |
Collapse
|
14
|
Talevi A, Bellera C. An update on the novel methods for the discovery of antiseizure and antiepileptogenic medications: where are we in 2024? Expert Opin Drug Discov 2024; 19:975-990. [PMID: 38963148 DOI: 10.1080/17460441.2024.2373165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Despite the availability of around 30 antiseizure medications, 1/3 of patients with epilepsy fail to become seizure-free upon pharmacological treatment. Available medications provide adequate symptomatic control in two-thirds of patients, but disease-modifying drugs are still scarce. Recently, though, new paradigms have been explored. AREAS COVERED Three areas are reviewed in which a high degree of innovation in the search for novel antiseizure and antiepileptogenic medications has been implemented: development of novel screening approaches, search for novel therapeutic targets, and adoption of new drug discovery paradigms aligned with a systems pharmacology perspective. EXPERT OPINION In the past, worldwide leaders in epilepsy have reiteratively stated that the lack of progress in the field may be explained by the recurrent use of the same molecular targets and screening procedures to identify novel medications. This landscape has changed recently, as reflected by the new Epilepsy Therapy Screening Program and the introduction of many in vitro and in vivo models that could possibly improve our chances of identifying first-in-class medications that may control drug-resistant epilepsy or modify the course of disease. Other milestones include the study of new molecular targets for disease-modifying drugs and exploration of a systems pharmacology perspective to design new drugs.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| | - Carolina Bellera
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| |
Collapse
|
15
|
Marotta N, Boland MJ, Prosser BL. Accelerating therapeutic development and clinical trial readiness for STXBP1 and SYNGAP1 disorders. Curr Probl Pediatr Adolesc Health Care 2024; 54:101576. [PMID: 38472035 DOI: 10.1016/j.cppeds.2024.101576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Gene-targeted therapies for genetic neurodevelopmental disorders (NDDs) are becoming a reality. The Center for Epilepsy and Neurodevelopmental Disorders (ENDD) is currently focused on the development of therapeutics for STXBP1 and SYNGAP1 disorders. Here we review the known clinical features of these disorders, highlight the biological role of STXBP1 and SYNGAP1, and discuss our current understanding of pathogenic mechanisms and therapeutic development. Finally, we provide our perspective as scientists and parents of children with NDDs, and comment on the current challenges for both clinical and basic science endeavors.
Collapse
Affiliation(s)
- Nicolas Marotta
- Department of Biochemistry and Molecular Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Center for Epilepsy and Neurodevelopmental Disorders (ENDD), University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael J Boland
- Center for Epilepsy and Neurodevelopmental Disorders (ENDD), University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Benjamin L Prosser
- Center for Epilepsy and Neurodevelopmental Disorders (ENDD), University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Physiology, Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Clark T, Waller MA, Loo L, Moreno CL, Denes CE, Neely GG. CRISPR activation screens: navigating technologies and applications. Trends Biotechnol 2024; 42:1017-1034. [PMID: 38493051 DOI: 10.1016/j.tibtech.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/18/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) activation (CRISPRa) has become an integral part of the molecular biology toolkit. CRISPRa genetic screens are an exciting high-throughput means of identifying genes the upregulation of which is sufficient to elicit a given phenotype. Activation machinery is continually under development to achieve greater, more robust, and more consistent activation. In this review, we offer a succinct technological overview of available CRISPRa architectures and a comprehensive summary of pooled CRISPRa screens. Furthermore, we discuss contemporary applications of CRISPRa across broad fields of research, with the aim of presenting a view of exciting emerging applications for CRISPRa screening.
Collapse
Affiliation(s)
- Teleri Clark
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Matthew A Waller
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Lipin Loo
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Cesar L Moreno
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Christopher E Denes
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - G Gregory Neely
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, and School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
17
|
Ye D, Chukwu C, Yang Y, Hu Z, Chen H. Adeno-associated virus vector delivery to the brain: Technology advancements and clinical applications. Adv Drug Deliv Rev 2024; 211:115363. [PMID: 38906479 DOI: 10.1016/j.addr.2024.115363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Adeno-associated virus (AAV) vectors have emerged as a promising tool in the development of gene therapies for various neurological diseases, including Alzheimer's disease and Parkinson's disease. However, the blood-brain barrier (BBB) poses a significant challenge to successfully delivering AAV vectors to the brain. Strategies that can overcome the BBB to improve the AAV delivery efficiency to the brain are essential to successful brain-targeted gene therapy. This review provides an overview of existing strategies employed for AAV delivery to the brain, including direct intraparenchymal injection, intra-cerebral spinal fluid injection, intranasal delivery, and intravenous injection of BBB-permeable AAVs. Focused ultrasound has emerged as a promising technology for the noninvasive and spatially targeted delivery of AAV administered by intravenous injection. This review also summarizes each strategy's current preclinical and clinical applications in treating neurological diseases. Moreover, this review includes a detailed discussion of the recent advances in the emerging focused ultrasound-mediated AAV delivery. Understanding the state-of-the-art of these gene delivery approaches is critical for future technology development to fulfill the great promise of AAV in neurological disease treatment.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Chinwendu Chukwu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Zhongtao Hu
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA; Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110 USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
18
|
Merolla A, Michetti C, Moschetta M, Vacca F, Ciano L, Emionite L, Astigiano S, Romei A, Horenkamp S, Berglund K, Gross RE, Cesca F, Colombo E, Benfenati F. A pH-sensitive closed-loop nanomachine to control hyperexcitability at the single neuron level. Nat Commun 2024; 15:5609. [PMID: 38965228 PMCID: PMC11224301 DOI: 10.1038/s41467-024-49941-3] [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: 09/28/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
Epilepsy affects 1% of the general population and 30% of patients are resistant to antiepileptic drugs. Although optogenetics is an efficient antiepileptic strategy, the difficulty of illuminating deep brain areas poses translational challenges. Thus, the search of alternative light sources is strongly needed. Here, we develop pH-sensitive inhibitory luminopsin (pHIL), a closed-loop chemo-optogenetic nanomachine composed of a luciferase-based light generator, a fluorescent sensor of intracellular pH (E2GFP), and an optogenetic actuator (halorhodopsin) for silencing neuronal activity. Stimulated by coelenterazine, pHIL experiences bioluminescence resonance energy transfer between luciferase and E2GFP which, under conditions of acidic pH, activates halorhodopsin. In primary neurons, pHIL senses the intracellular pH drop associated with hyperactivity and optogenetically aborts paroxysmal activity elicited by the administration of convulsants. The expression of pHIL in hippocampal pyramidal neurons is effective in decreasing duration and increasing latency of pilocarpine-induced tonic-clonic seizures upon in vivo coelenterazine administration, without affecting higher brain functions. The same treatment is effective in markedly decreasing seizure manifestations in a murine model of genetic epilepsy. The results indicate that pHIL represents a potentially promising closed-loop chemo-optogenetic strategy to treat drug-refractory epilepsy.
Collapse
Affiliation(s)
- Assunta Merolla
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Matteo Moschetta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Vacca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Lorenzo Ciano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Alessandra Romei
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Simone Horenkamp
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
19
|
Veltra D, Theodorou V, Katsalouli M, Vorgia P, Niotakis G, Tsaprouni T, Pons R, Kosma K, Kampouraki A, Tsoutsou I, Makrythanasis P, Kekou K, Traeger-Synodinos J, Sofocleous C. SCN1A Channels a Wide Range of Epileptic Phenotypes: Report of Novel and Known Variants with Variable Presentations. Int J Mol Sci 2024; 25:5644. [PMID: 38891831 PMCID: PMC11171476 DOI: 10.3390/ijms25115644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
SCN1A, the gene encoding for the Nav1.1 channel, exhibits dominant interneuron-specific expression, whereby variants disrupting the channel's function affect the initiation and propagation of action potentials and neuronal excitability causing various types of epilepsy. Dravet syndrome (DS), the first described clinical presentation of SCN1A channelopathy, is characterized by severe myoclonic epilepsy in infancy (SMEI). Variants' characteristics and other genetic or epigenetic factors lead to extreme clinical heterogeneity, ranging from non-epileptic conditions to developmental and epileptic encephalopathy (DEE). This current study reports on findings from 343 patients referred by physicians in hospitals and tertiary care centers in Greece between 2017 and 2023. Positive family history for specific neurologic disorders was disclosed in 89 cases and the one common clinical feature was the onset of seizures, at a mean age of 17 months (range from birth to 15 years old). Most patients were specifically referred for SCN1A investigation (Sanger Sequencing and MLPA) and only five for next generation sequencing. Twenty-six SCN1A variants were detected, including nine novel causative variants (c.4567A>Τ, c.5564C>A, c.2176+2T>C, c.3646G>C, c.4331C>A, c.1130_1131delGAinsAC, c.1574_1580delCTGAGGA, c.4620A>G and c.5462A>C), and are herein presented, along with subsequent genotype-phenotype associations. The identification of novel variants complements SCN1A databases extending our expertise on genetic counseling and patient and family management including gene-based personalized interventions.
Collapse
Affiliation(s)
- Danai Veltra
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
- Research University Institute for the Study and Prevention of Genetic and Malignant Disease of Childhood, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece
| | - Virginia Theodorou
- Pediatric Neurology Department, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (V.T.); (M.K.)
| | - Marina Katsalouli
- Pediatric Neurology Department, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (V.T.); (M.K.)
| | - Pelagia Vorgia
- Agrifood and Life Sciences Institute, Hellenic Mediterranean University, 71410 Heraklion, Greece;
| | - Georgios Niotakis
- Pediatric Neurology Department, Venizelion Hospital, 71409 Heraklion, Greece;
| | | | - Roser Pons
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece;
| | - Konstantina Kosma
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| | - Afroditi Kampouraki
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| | - Irene Tsoutsou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| | - Periklis Makrythanasis
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
- Department of Genetic Medicine and Development, Medical School, University of Geneva, 1211 Geneva, Switzerland
- Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Kyriaki Kekou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| | - Joanne Traeger-Synodinos
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| | - Christalena Sofocleous
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, St. Sophia’s Children’s Hospital, 11527 Athens, Greece; (D.V.); (K.K.); (A.K.); (I.T.); (P.M.); (K.K.); (J.T.-S.)
| |
Collapse
|
20
|
Di Berardino C, Mainardi M, Brusco S, Benvenuto E, Broccoli V, Colasante G. Temporal manipulation of the Scn1a gene reveals its essential role in adult brain function. Brain 2024; 147:1216-1230. [PMID: 37812819 PMCID: PMC10994529 DOI: 10.1093/brain/awad350] [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: 05/23/2023] [Revised: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023] Open
Abstract
Dravet syndrome is a severe epileptic encephalopathy, characterized by drug-resistant epilepsy, severe cognitive and behavioural deficits, with increased risk of sudden unexpected death (SUDEP). It is caused by haploinsufficiency of SCN1A gene encoding for the α-subunit of the voltage-gated sodium channel Nav1.1. Therapeutic approaches aiming to upregulate the healthy copy of SCN1A gene to restore its normal expression levels are being developed. However, whether Scn1a gene function is required only during a specific developmental time-window or, alternatively, if its physiological expression is necessary in adulthood is untested up to now. We induced Scn1a gene haploinsufficiency at two ages spanning postnatal brain development (P30 and P60) and compared the phenotypes of those mice to Scn1a perinatally induced mice (P2), recapitulating all deficits of Dravet mice. Induction of heterozygous Nav1.1 mutation at P30 and P60 elicited susceptibility to the development of both spontaneous and hyperthermia-induced seizures and SUDEP rates comparable to P2-induced mice, with symptom onset accompanied by the characteristic GABAergic interneuron dysfunction. Finally, delayed Scn1a haploinsufficiency induction provoked hyperactivity, anxiety and social attitude impairment at levels comparable to age matched P2-induced mice, while it was associated with a better cognitive performance, with P60-induced mice behaving like the control group. Our data show that maintenance of physiological levels of Nav1.1 during brain development is not sufficient to prevent Dravet symptoms and that long-lasting restoration of Scn1a gene expression would be required to grant optimal clinical benefit in patients with Dravet syndrome.
Collapse
Affiliation(s)
- Claudia Di Berardino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Martina Mainardi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Simone Brusco
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy
| | - Elena Benvenuto
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Gene and Cell Therapy PhD Program, Vita- Salute San Raffaele University, 20132 Milan, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
21
|
Nelson AD, Catalfio AM, Gupta JP, Min L, Caballero-Florán RN, Dean KP, Elvira CC, Derderian KD, Kyoung H, Sahagun A, Sanders SJ, Bender KJ, Jenkins PM. Physical and functional convergence of the autism risk genes Scn2a and Ank2 in neocortical pyramidal cell dendrites. Neuron 2024; 112:1133-1149.e6. [PMID: 38290518 PMCID: PMC11097922 DOI: 10.1016/j.neuron.2024.01.003] [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: 06/08/2022] [Revised: 04/26/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
Dysfunction in sodium channels and their ankyrin scaffolding partners have both been implicated in neurodevelopmental disorders, including autism spectrum disorder (ASD). In particular, the genes SCN2A, which encodes the sodium channel NaV1.2, and ANK2, which encodes ankyrin-B, have strong ASD association. Recent studies indicate that ASD-associated haploinsufficiency in Scn2a impairs dendritic excitability and synaptic function in neocortical pyramidal cells, but how NaV1.2 is anchored within dendritic regions is unknown. Here, we show that ankyrin-B is essential for scaffolding NaV1.2 to the dendritic membrane of mouse neocortical neurons and that haploinsufficiency of Ank2 phenocopies intrinsic dendritic excitability and synaptic deficits observed in Scn2a+/- conditions. These results establish a direct, convergent link between two major ASD risk genes and reinforce an emerging framework suggesting that neocortical pyramidal cell dendritic dysfunction can contribute to neurodevelopmental disorder pathophysiology.
Collapse
Affiliation(s)
- Andrew D Nelson
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Amanda M Catalfio
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Julie P Gupta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lia Min
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kendall P Dean
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carina C Elvira
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kimberly D Derderian
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Kyoung
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Atehsa Sahagun
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephan J Sanders
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin J Bender
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Paul M Jenkins
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Chrzanowski S, Batra R. CRISPR-Based Gene Editing Techniques in Pediatric Neurological Disorders. Pediatr Neurol 2024; 153:166-174. [PMID: 38394831 DOI: 10.1016/j.pediatrneurol.2024.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
The emergence of gene editing technologies offers a unique opportunity to develop mutation-specific treatments for pediatric neurological disorders. Gene editing systems can potentially alter disease trajectory by correcting dysfunctional mutations or therapeutically altering gene expression. Clustered regularly interspaced short palindromic repeats (CRISPR)-based approaches are attractive gene therapy platforms to personalize treatments because of their specificity, ease of design, versatility, and cost. However, many such approaches remain in the early stages of development, with ongoing efforts to optimize editing efficiency, minimize unintended off-target effects, and mitigate pathologic immune responses. Given the rapid evolution of CRISPR-based therapies, it is prudent for the clinically based child neurologist to have a conceptual understanding of what such therapies may entail, including both benefits and risks and how such therapies may be clinically applied. In this review, we describe the fundamentals of CRISPR-based therapies, discuss the opportunities and challenges that have arisen, and highlight preclinical work in several pediatric neurological diseases.
Collapse
Affiliation(s)
- Stephen Chrzanowski
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts; Division of Neuromuscular Medicine, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts; Division of Neuromuscular Medicine, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts.
| | | |
Collapse
|
23
|
Chen GT, Nair G, Osorio AJ, Holley SM, Ghassemzadeh K, Gonzalez J, Lu C, Sanjana NE, Cepeda C, Geschwind DH. Enhancer-targeted CRISPR-Activation Rescues Haploinsufficient Autism Susceptibility Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584921. [PMID: 38559217 PMCID: PMC10980046 DOI: 10.1101/2024.03.13.584921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Autism Spectrum Disorder (ASD) is a highly heritable condition with diverse clinical presentations. Approximately 20% of ASD's genetic susceptibility is imparted by de novo mutations of major effect, most of which cause haploinsufficiency. We mapped enhancers of two high confidence autism genes - CHD8 and SCN2A and used CRISPR-based gene activation (CRISPR-A) in hPSC-derived excitatory neurons and cerebral forebrain organoids to correct the effects of haploinsufficiency, taking advantage of the presence of a wildtype allele of each gene and endogenous gene regulation. We found that CRISPR-A induced a sustained increase in CHD8 and SCN2A expression in treated neurons and organoids, with rescue of gene expression levels and mutation-associated phenotypes, including gene expression and physiology. These data support gene activation via targeting enhancers of haploinsufficient genes, as a therapeutic intervention in ASD and other neurodevelopmental disorders.
Collapse
|
24
|
Jiang H, Tang M, Xu Z, Wang Y, Li M, Zheng S, Zhu J, Lin Z, Zhang M. CRISPR/Cas9 system and its applications in nervous system diseases. Genes Dis 2024; 11:675-686. [PMID: 37692518 PMCID: PMC10491921 DOI: 10.1016/j.gendis.2023.03.017] [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: 11/28/2022] [Accepted: 03/05/2023] [Indexed: 09/12/2023] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system is an acquired immune system of many bacteria and archaea, comprising CRISPR loci, Cas genes, and its associated proteins. This system can recognize exogenous DNA and utilize the Cas9 protein's nuclease activity to break DNA double-strand and to achieve base insertion or deletion by subsequent DNA repair. In recent years, multiple laboratory and clinical studies have revealed the therapeutic role of the CRISPR/Cas9 system in neurological diseases. This article reviews the CRISPR/Cas9-mediated gene editing technology and its potential for clinical application against neurological diseases.
Collapse
Affiliation(s)
- Haibin Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengyan Tang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zidi Xu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yanan Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mopu Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shuyin Zheng
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianghu Zhu
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| | - Min Zhang
- Department of Pediatrics, The Second School of Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang 325000, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
25
|
Bettegazzi B, Cattaneo S, Simonato M, Zucchini S, Soukupova M. Viral Vector-Based Gene Therapy for Epilepsy: What Does the Future Hold? Mol Diagn Ther 2024; 28:5-13. [PMID: 38103141 PMCID: PMC10786988 DOI: 10.1007/s40291-023-00687-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2023] [Indexed: 12/17/2023]
Abstract
In recent years, many pre-clinical studies have tested gene therapy approaches as possible treatments for epilepsy, following the idea that they may provide an alternative to conventional pharmacological and surgical options. Multiple gene therapy approaches have been developed, including those based on anti-sense oligonucleotides, RNA interference, and viral vectors. In this opinion article, we focus on translational issues related to viral vector-mediated gene therapy for epilepsy. Research has advanced dramatically in addressing issues like viral vector optimization, target identification, strategies of gene expression, editing or regulation, and safety. Some of these pre-clinically validated potential gene therapies are now being tested in clinical trials, in patients with genetic or focal forms of drug-resistant epilepsy. Here, we discuss the ongoing translational research and the advancements that are needed and expected in the near future. We then describe the clinical trials in the pipeline and the further challenges that will need to be addressed at the clinical and economic levels. Our optimistic view is that all these issues and challenges can be overcome, and that gene therapy approaches for epilepsy will soon become a clinical reality.
Collapse
Affiliation(s)
| | - Stefano Cattaneo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Michele Simonato
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| | - Silvia Zucchini
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy.
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, Ferrara, Italy.
| | - Marie Soukupova
- Department of Neuroscience and Rehabilitation, University of Ferrara, via Fossato di Mortara 70, 44121, Ferrara, Italy
| |
Collapse
|
26
|
Fan HC, Yang MT, Lin LC, Chiang KL, Chen CM. Clinical and Genetic Features of Dravet Syndrome: A Prime Example of the Role of Precision Medicine in Genetic Epilepsy. Int J Mol Sci 2023; 25:31. [PMID: 38203200 PMCID: PMC10779156 DOI: 10.3390/ijms25010031] [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: 11/23/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Dravet syndrome (DS), also known as severe myoclonic epilepsy of infancy, is a rare and drug-resistant form of developmental and epileptic encephalopathies, which is both debilitating and challenging to manage, typically arising during the first year of life, with seizures often triggered by fever, infections, or vaccinations. It is characterized by frequent and prolonged seizures, developmental delays, and various other neurological and behavioral impairments. Most cases result from pathogenic mutations in the sodium voltage-gated channel alpha subunit 1 (SCN1A) gene, which encodes a critical voltage-gated sodium channel subunit involved in neuronal excitability. Precision medicine offers significant potential for improving DS diagnosis and treatment. Early genetic testing enables timely and accurate diagnosis. Advances in our understanding of DS's underlying genetic mechanisms and neurobiology have enabled the development of targeted therapies, such as gene therapy, offering more effective and less invasive treatment options for patients with DS. Targeted and gene therapies provide hope for more effective and personalized treatments. However, research into novel approaches remains in its early stages, and their clinical application remains to be seen. This review addresses the current understanding of clinical DS features, genetic involvement in DS development, and outcomes of novel DS therapies.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan;
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Ming-Tao Yang
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 320, Taiwan
| | - Lung-Chang Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Kuo-Liang Chiang
- Department of Pediatric Neurology, Kuang-Tien General Hospital, Taichung 433, Taiwan;
- Department of Nutrition, Hungkuang University, Taichung 433, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
27
|
Mich JK, Ryu J, Wei AD, Gore BB, Guo R, Bard AM, Martinez RA, Bishaw Y, Luber E, Oliveira Santos LM, Miranda N, Ramirez JM, Ting JT, Lein ES, Levi BP, Kalume FK. AAV-mediated interneuron-specific gene replacement for Dravet syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571820. [PMID: 38168178 PMCID: PMC10760176 DOI: 10.1101/2023.12.15.571820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Dravet syndrome (DS) is a devastating developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10-20% rate of premature death. Most DS patients harbor loss-of-function mutations in one copy of SCN1A , which has been associated with inhibitory neuron dysfunction. Here we developed an interneuron-targeting AAV human SCN1A gene replacement therapy using cell class-specific enhancers. We generated a split-intein fusion form of SCN1A to circumvent AAV packaging limitations and deliver SCN1A via a dual vector approach using cell class-specific enhancers. These constructs produced full-length Na V 1.1 protein and functional sodium channels in HEK293 cells and in brain cells in vivo . After packaging these vectors into enhancer-AAVs and administering to mice, immunohistochemical analyses showed telencephalic GABAergic interneuron-specific and dose-dependent transgene biodistribution. These vectors conferred strong dose-dependent protection against postnatal mortality and seizures in two DS mouse models carrying independent loss-of-function alleles of Scn1a, at two independent research sites, supporting the robustness of this approach. No mortality or toxicity was observed in wild-type mice injected with single vectors expressing either the N-terminal or C-terminal halves of SCN1A , or the dual vector system targeting interneurons. In contrast, nonselective neuronal targeting of SCN1A conferred less rescue against mortality and presented substantial preweaning lethality. These findings demonstrate proof-of-concept that interneuron-specific AAV-mediated SCN1A gene replacement is sufficient for significant rescue in DS mouse models and suggest it could be an effective therapeutic approach for patients with DS.
Collapse
|
28
|
Ricobaraza A, Bunuales M, Gonzalez-Aparicio M, Fadila S, Rubinstein M, Vides-Urrestarazu I, Banderas J, Sola-Sevilla N, Sanchez-Carpintero R, Lanciego JL, Roda E, Honrubia A, Arnaiz P, Hernandez-Alcoceba R. Preferential expression of SCN1A in GABAergic neurons improves survival and epileptic phenotype in a mouse model of Dravet syndrome. J Mol Med (Berl) 2023; 101:1587-1601. [PMID: 37819378 PMCID: PMC10697872 DOI: 10.1007/s00109-023-02383-8] [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: 05/05/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
The SCN1A gene encodes the alpha subunit of a voltage-gated sodium channel (Nav1.1), which is essential for the function of inhibitory neurons in the brain. Mutations in this gene cause severe encephalopathies such as Dravet syndrome (DS). Upregulation of SCN1A expression by different approaches has demonstrated promising therapeutic effects in preclinical models of DS. Limiting the effect to inhibitory neurons may contribute to the restoration of brain homeostasis, increasing the safety and efficacy of the treatment. In this work, we have evaluated different approaches to obtain preferential expression of the full SCN1A cDNA (6 Kb) in GABAergic neurons, using high-capacity adenoviral vectors (HC-AdV). In order to favour infection of these cells, we considered ErbB4 as a surface target. Incorporation of the EGF-like domain from neuregulin 1 alpha (NRG1α) in the fiber of adenovirus capsid allowed preferential infection in cells lines expressing ErbB4. However, it had no impact on the infectivity of the vector in primary cultures or in vivo. For transcriptional control of transgene expression, we developed a regulatory sequence (DP3V) based on the Distal-less homolog enhancer (Dlx), the vesicular GABA transporter (VGAT) promoter, and a portion of the SCN1A gene. The hybrid DP3V promoter allowed preferential expression of transgenes in GABAergic neurons both in vitro and in vivo. A new HC-AdV expressing SCN1A under the control of this promoter showed improved survival and amelioration of the epileptic phenotype in a DS mouse model. These results increase the repertoire of gene therapy vectors for the treatment of DS and indicate a new avenue for the refinement of gene supplementation in this disease. KEY MESSAGES: Adenoviral vectors can deliver the SCN1A cDNA and are amenable for targeting. An adenoviral vector displaying an ErbB4 ligand in the capsid does not target GABAergic neurons. A hybrid promoter allows preferential expression of transgenes in GABAergic neurons. Preferential expression of SCN1A in GABAergic cells is therapeutic in a Dravet syndrome model.
Collapse
Affiliation(s)
- Ana Ricobaraza
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Maria Bunuales
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Manuela Gonzalez-Aparicio
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Saja Fadila
- Sackler Faculty of Medicine, Goldschleger Eye Research Institute, Tel Aviv University, Tel Aviv, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moran Rubinstein
- Sackler Faculty of Medicine, Goldschleger Eye Research Institute, Tel Aviv University, Tel Aviv, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irene Vides-Urrestarazu
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Julliana Banderas
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Noemi Sola-Sevilla
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain
| | - Rocio Sanchez-Carpintero
- University Clinic of Navarra, Dravet Syndrome Unit, Pediatric Neurology Unit, IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Jose Luis Lanciego
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Madrid, Spain
| | - Elvira Roda
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Adriana Honrubia
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Patricia Arnaiz
- Department of Neuroscience, CIMA, University of Navarra, Pamplona, Spain
| | - Ruben Hernandez-Alcoceba
- Gene Therapy and Regulation of Gene Expression Program, CIMA, University of Navarra, CIMA, Av. Pio XII 55, E-31008, Pamplona, Spain.
| |
Collapse
|
29
|
Lersch R, Jannadi R, Grosse L, Wagner M, Schneider MF, von Stülpnagel C, Heinen F, Potschka H, Borggraefe I. Targeted Molecular Strategies for Genetic Neurodevelopmental Disorders: Emerging Lessons from Dravet Syndrome. Neuroscientist 2023; 29:732-750. [PMID: 35414300 PMCID: PMC10623613 DOI: 10.1177/10738584221088244] [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: 11/17/2022]
Abstract
Dravet syndrome is a severe developmental and epileptic encephalopathy mostly caused by heterozygous mutation of the SCN1A gene encoding the voltage-gated sodium channel α subunit Nav1.1. Multiple seizure types, cognitive deterioration, behavioral disturbances, ataxia, and sudden unexpected death associated with epilepsy are a hallmark of the disease. Recently approved antiseizure medications such as fenfluramine and cannabidiol have been shown to reduce seizure burden. However, patients with Dravet syndrome are still medically refractory in the majority of cases, and there is a high demand for new therapies aiming to improve behavioral and cognitive outcome. Drug-repurposing approaches for SCN1A-related Dravet syndrome are currently under investigation (i.e., lorcaserin, clemizole, and ataluren). New therapeutic concepts also arise from the field of precision medicine by upregulating functional SCN1A or by activating Nav1.1. These include antisense nucleotides directed against the nonproductive transcript of SCN1A with the poison exon 20N and against an inhibitory noncoding antisense RNA of SCN1A. Gene therapy approaches such as adeno-associated virus-based upregulation of SCN1A using a transcriptional activator (ETX101) or CRISPR/dCas technologies show promising results in preclinical studies. Although these new treatment concepts still need further clinical research, they offer great potential for precise and disease modifying treatment of Dravet syndrome.
Collapse
Affiliation(s)
- Robert Lersch
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Rawan Jannadi
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
- Institute of Human Genetics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Leonie Grosse
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Matias Wagner
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute for Neurogenomics, Helmholtz Centre Munich, German Research Center for Health and Environment (GmbH), Munich, Germany
| | - Marius Frederik Schneider
- Metabolic Biochemistry, Biomedical Center Munich, Medical Faculty, Ludwig Maximilians University, Munich, Germany
- International Max Planck Research School (IMPRS) for Molecular Life Sciences, Planegg-Martinsried, Germany
| | - Celina von Stülpnagel
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
- Research Institute for Rehabilitation, Transition and Palliation, Paracelsus Medical Private University (PMU), Salzburg, Austria
| | - Florian Heinen
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilians University, Munich, Germany
| | - Ingo Borggraefe
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
- Comprehensive Epilepsy Center, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
30
|
Whyte-Fagundes P, Vance A, Carroll A, Figueroa F, Manukyan C, Baraban SC. Testing of putative antiseizure drugs in a preclinical Dravet syndrome zebrafish model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566723. [PMID: 38014342 PMCID: PMC10680609 DOI: 10.1101/2023.11.11.566723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Dravet syndrome (DS) is a severe genetic epilepsy primarily caused by de novo mutations in a voltage-activated sodium channel gene (SCN1A). Patients face life-threatening seizures that are largely resistant to available anti-seizure medications (ASM). Preclinical DS animal models are a valuable tool to identify candidate ASMs for these patients. Among these, scn1lab mutant zebrafish exhibiting spontaneous seizure-like activity are particularly amenable to large-scale drug screening. Prior screening in a scn1lab mutant zebrafish line generated using N-ethyl-Nnitrosourea (ENU) identified valproate, stiripentol, and fenfluramine e.g., Federal Drug Administration (FDA) approved drugs with clinical application in the DS population. Successful phenotypic screening in scn1lab mutant zebrafish consists of two stages: (i) a locomotion-based assay measuring high-velocity convulsive swim behavior and (ii) an electrophysiology-based assay, using in vivo local field potential (LFP) recordings, to quantify electrographic seizure-like events. Using this strategy more than 3000 drug candidates have been screened in scn1lab zebrafish mutants. Here, we curated a list of nine additional anti-seizure drug candidates recently identified in preclinical models: 1-EBIO, AA43279, chlorzoxazone, donepezil, lisuride, mifepristone, pargyline, soticlestat and vorinostat. First-stage locomotion-based assays in scn1lab mutant zebrafish identified only 1-EBIO, chlorzoxazone and lisuride. However, second-stage LFP recording assays did not show significant suppression of spontaneous electrographic seizure activity for any of the nine anti-seizure drug candidates. Surprisingly, soticlestat induced frank electrographic seizure-like discharges in wild-type control zebrafish. Taken together, our results failed to replicate clear anti-seizure efficacy for these drug candidates highlighting a necessity for strict scientific standards in preclinical identification of ASMs.
Collapse
Affiliation(s)
- P Whyte-Fagundes
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - A Vance
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - A Carroll
- Behavioral Neurosciences, Northeastern University, Boston, MA, USA
| | - F Figueroa
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - C Manukyan
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - S C Baraban
- Epilepsy Research Laboratory and Weill Institute for Neuroscience, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
31
|
Song C, Zhao Y, Zhang J, Dong Z, Kang X, Pan Y, Du J, Gao Y, Zhang H, Xi Y, Ding H, Kuang F, Wang W, Luo C, Zhang Z, Zhao Q, Yang J, Jiang W, Wu S, Gao F. Spatial Distribution of Parvalbumin-Positive Fibers in the Mouse Brain and Their Alterations in Mouse Models of Temporal Lobe Epilepsy and Parkinson's Disease. Neurosci Bull 2023; 39:1683-1702. [PMID: 37523099 PMCID: PMC10603013 DOI: 10.1007/s12264-023-01083-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 03/27/2023] [Indexed: 08/01/2023] Open
Abstract
Parvalbumin interneurons belong to the major types of GABAergic interneurons. Although the distribution and pathological alterations of parvalbumin interneuron somata have been widely studied, the distribution and vulnerability of the neurites and fibers extending from parvalbumin interneurons have not been detailly interrogated. Through the Cre recombinase-reporter system, we visualized parvalbumin-positive fibers and thoroughly investigated their spatial distribution in the mouse brain. We found that parvalbumin fibers are widely distributed in the brain with specific morphological characteristics in different regions, among which the cortex and thalamus exhibited the most intense parvalbumin signals. In regions such as the striatum and optic tract, even long-range thick parvalbumin projections were detected. Furthermore, in mouse models of temporal lobe epilepsy and Parkinson's disease, parvalbumin fibers suffered both massive and subtle morphological alterations. Our study provides an overview of parvalbumin fibers in the brain and emphasizes the potential pathological implications of parvalbumin fiber alterations.
Collapse
Affiliation(s)
- Changgeng Song
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Zhao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiajia Zhang
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ziyi Dong
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Kang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuqi Pan
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinle Du
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiting Gao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Haifeng Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ye Xi
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Ding
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Fang Kuang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenting Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhengping Zhang
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710054, China
| | - Qinpeng Zhao
- Department of Spinal Surgery, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, 710054, China
| | - Jiazhou Yang
- The Medical College of Yan'an University, Yan'an, 716000, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
32
|
Cao BR, Huang YM, Tian FY, Li JH, Xu CL, Wei Y, Liu JK, Guo QB, Xu HY, Zhan L, Lv RM, Sun YD, Hu XD, Gao ZB, Zhou CY. Prime editing-based gene correction alleviates the hyperexcitable phenotype and seizures of a genetic epilepsy mouse model. Acta Pharmacol Sin 2023; 44:2342-2345. [PMID: 37402996 PMCID: PMC10618215 DOI: 10.1038/s41401-023-01115-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/21/2023] [Indexed: 07/06/2023] Open
Affiliation(s)
- Bi-Rong Cao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi-Ming Huang
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fu-Yun Tian
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Jin-Hui Li
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chun-Long Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Wei
- Shanghai Immunocan Biotech Co., LTD, Shanghai, 201800, China
| | - Ji-Kai Liu
- Shanghai Immunocan Biotech Co., LTD, Shanghai, 201800, China
| | - Qian-Bei Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai-Yan Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Zhan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Rui-Min Lv
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi-di Sun
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xin-de Hu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhao-Bing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Chang-Yang Zhou
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
33
|
Riedmayr LM, Hinrichsmeyer KS, Thalhammer SB, Mittas DM, Karguth N, Otify DY, Böhm S, Weber VJ, Bartoschek MD, Splith V, Brümmer M, Ferreira R, Boon N, Wögenstein GM, Grimm C, Wijnholds J, Mehlfeld V, Michalakis S, Fenske S, Biel M, Becirovic E. mRNA trans-splicing dual AAV vectors for (epi)genome editing and gene therapy. Nat Commun 2023; 14:6578. [PMID: 37852949 PMCID: PMC10584818 DOI: 10.1038/s41467-023-42386-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/10/2023] [Indexed: 10/20/2023] Open
Abstract
Large genes including several CRISPR-Cas modules like gene activators (CRISPRa) require dual adeno-associated viral (AAV) vectors for an efficient in vivo delivery and expression. Current dual AAV vector approaches have important limitations, e.g., low reconstitution efficiency, production of alien proteins, or low flexibility in split site selection. Here, we present a dual AAV vector technology based on reconstitution via mRNA trans-splicing (REVeRT). REVeRT is flexible in split site selection and can efficiently reconstitute different split genes in numerous in vitro models, in human organoids, and in vivo. Furthermore, REVeRT can functionally reconstitute a CRISPRa module targeting genes in various mouse tissues and organs in single or multiplexed approaches upon different routes of administration. Finally, REVeRT enabled the reconstitution of full-length ABCA4 after intravitreal injection in a mouse model of Stargardt disease. Due to its flexibility and efficiency REVeRT harbors great potential for basic research and clinical applications.
Collapse
Affiliation(s)
- Lisa Maria Riedmayr
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | | | | | - David Manuel Mittas
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Nina Karguth
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Dina Yehia Otify
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | | | - Valentin Johannes Weber
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | | | | | - Manuela Brümmer
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Raphael Ferreira
- Genetics Department, Harvard Medical School, Boston, MA, 02115, USA
| | - Nanda Boon
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA, Leiden, Netherlands
| | - Gabriele Maria Wögenstein
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA, Leiden, Netherlands
- Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA, Amsterdam, Netherlands
| | - Verena Mehlfeld
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80336, Munich, Germany
| | - Stefanie Fenske
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, 81377, Germany
| | - Martin Biel
- Department of Pharmacy - Center for Drug Research, LMU Munich, Munich, 81377, Germany
| | - Elvir Becirovic
- Laboratory for Retinal Gene Therapy, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Schlieren, 8952, Switzerland.
| |
Collapse
|
34
|
Griazeva ED, Fedoseeva DM, Radion EI, Ershov PV, Meshkov IO, Semyanihina AV, Makarova AS, Makarov VV, Yudin VS, Keskinov AA, Kraevoy SA. Current Approaches to Epigenetic Therapy. EPIGENOMES 2023; 7:23. [PMID: 37873808 PMCID: PMC10594535 DOI: 10.3390/epigenomes7040023] [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: 08/24/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 10/25/2023] Open
Abstract
Epigenetic therapy is a promising tool for the treatment of a wide range of diseases. Several fundamental epigenetic approaches have been proposed. Firstly, the use of small molecules as epigenetic effectors, as the most developed pharmacological method, has contributed to the introduction of a number of drugs into clinical practice. Secondly, various innovative epigenetic approaches based on dCas9 and the use of small non-coding RNAs as therapeutic agents are also under extensive research. In this review, we present the current state of research in the field of epigenetic therapy, considering the prospects for its application and possible limitations.
Collapse
Affiliation(s)
- Ekaterina D. Griazeva
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Daria M. Fedoseeva
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Elizaveta I. Radion
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Pavel V. Ershov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Ivan O. Meshkov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Alexandra V. Semyanihina
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
- Federal State Budgetary Institution “N.N. Blokhin National Medical Research Center of Oncology” of the Ministry of Health of the Russian Federation (N.N. Blokhin NMRCO), Kashirskoe Shosse, 24, Moscow 115478, Russia
- Federal State Budgetary Scientific Institution, Research Centre for Medical Genetics, Moskvorechye, 1, Moscow 115522, Russia
| | - Anna S. Makarova
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Valentin V. Makarov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Vladimir S. Yudin
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Anton A. Keskinov
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| | - Sergey A. Kraevoy
- Federal State Budgetary Institution, Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Pogodinskaya Str., 10, Building 1, Moscow 119121, Russia
| |
Collapse
|
35
|
Kioutchoukova IP, Foster DT, Thakkar RN, Foreman MA, Burgess BJ, Toms RM, Molina Valero EE, Lucke-Wold B. Neurologic orphan diseases: Emerging innovations and role for genetic treatments. World J Exp Med 2023; 13:59-74. [PMID: 37767543 PMCID: PMC10520757 DOI: 10.5493/wjem.v13.i4.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/16/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023] Open
Abstract
Orphan diseases are rare diseases that affect less than 200000 individuals within the United States. Most orphan diseases are of neurologic and genetic origin. With the current advances in technology, more funding has been devoted to developing therapeutic agents for patients with these conditions. In our review, we highlight emerging options for patients with neurologic orphan diseases, specifically including diseases resulting in muscular deterioration, epilepsy, seizures, neurodegenerative movement disorders, inhibited cognitive development, neuron deterioration, and tumors. After extensive literature review, gene therapy offers a promising route for the treatment of neurologic orphan diseases. The use of clustered regularly interspaced palindromic repeats/Cas9 has demonstrated positive results in experiments investigating its role in several diseases. Additionally, the use of adeno-associated viral vectors has shown improvement in survival, motor function, and developmental milestones, while also demonstrating reversal of sensory ataxia and cardiomyopathy in Friedreich ataxia patients. Antisense oligonucleotides have also been used in some neurologic orphan diseases with positive outcomes. Mammalian target of rapamycin inhibitors are currently being investigated and have reduced abnormal cell growth, proliferation, and angiogenesis. Emerging innovations and the role of genetic treatments open a new window of opportunity for the treatment of neurologic orphan diseases.
Collapse
Affiliation(s)
| | - Devon T Foster
- Florida International University Herbert Wertheim College of Medicine, Florida International University Herbert Wertheim College of Medicine, Miami, FL 33199, United States
| | - Rajvi N Thakkar
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Marco A Foreman
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Brandon J Burgess
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Rebecca M Toms
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
36
|
Brentari I, Zadorozhna M, Denti MA, Giorgio E. RNA therapeutics for neurological diseases. Br Med Bull 2023; 147:50-61. [PMID: 37210633 DOI: 10.1093/bmb/ldad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/27/2023] [Accepted: 05/09/2023] [Indexed: 05/22/2023]
Abstract
INTRODUCTION Ribonucleic acid (RNA) therapeutics are a new class of drugs whose importance is highlighted by the growing number of molecules in the clinic. SOURCES OF DATA We focus on RNA therapeutics for neurogenetic disorders, which are broadly defined as diseases with a genetic background and with at least one clinical sign affecting the nervous system. A systematic search identified 14 RNA drugs approved by FDA and many others in development. AREAS OF AGREEMENT The field of RNA therapeutics is changing the therapeutic scenario across many disorders. AREAS OF CONTROVERSY Despite its recent successes, RNA therapeutics encountered several hurdles and some clinical failures. Delivery to the brain represents the biggest challenge. GROWING POINTS The many advantages of RNA drugs make the development of these technologies a worthwhile investment. AREAS TIMELY FOR DEVELOPING RESEARCH Clinical failures stress the importance of implementing clinical trial design and optimizing RNA molecules to hold the promise of revolutionizing the treatment of human diseases.
Collapse
Affiliation(s)
- Ilaria Brentari
- Dipartimento di Biologia Cellulare, Computazionale e Integrata (CIBIO), Università degli Studi di Trento, 38123 Trento, Italy
| | - Mariia Zadorozhna
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Michela Alessandra Denti
- Dipartimento di Biologia Cellulare, Computazionale e Integrata (CIBIO), Università degli Studi di Trento, 38123 Trento, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Medical Genetics Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy
| |
Collapse
|
37
|
Lopez L, De Waard S, Meudal H, Caumes C, Khakh K, Peigneur S, Oliveira-Mendes B, Lin S, De Waele J, Montnach J, Cestèle S, Tessier A, Johnson JP, Mantegazza M, Tytgat J, Cohen C, Béroud R, Bosmans F, Landon C, De Waard M. Structure-function relationship of new peptides activating human Na v1.1. Biomed Pharmacother 2023; 165:115173. [PMID: 37453200 DOI: 10.1016/j.biopha.2023.115173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Nav1.1 is an important pharmacological target as this voltage-gated sodium channel is involved in neurological and cardiac syndromes. Channel activators are actively sought to try to compensate for haploinsufficiency in several of these pathologies. Herein we used a natural source of new peptide compounds active on ion channels and screened for drugs capable to inhibit channel inactivation as a way to compensate for decreased channel function. We discovered that JzTx-34 is highly active on Nav1.1 and subsequently performed a full structure-activity relationship investigation to identify its pharmacophore. These experiments will help interpret the mechanism of action of this and formerly identified peptides as well as the future identification of new peptides. We also reveal structural determinants that make natural ICK peptides active against Nav1.1 challenging to synthesize. Altogether, the knowledge gained by this study will help facilitate the discovery and development of new compounds active on this critical ion channel target.
Collapse
Affiliation(s)
- Ludivine Lopez
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Smartox Biotechnology, Saint-Egrève, France
| | - Stephan De Waard
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; LabEx "Ion Channels, Science and Therapeutics", Valbonne, France
| | - Hervé Meudal
- Center for Molecular Biophysics, CNRS, rue Charles Sadron, CS 80054, Orléans 45071, France
| | | | - Kuldip Khakh
- Xenon Pharmaceuticals, Burnaby, British Columbia, Canada
| | | | | | - Sophia Lin
- Xenon Pharmaceuticals, Burnaby, British Columbia, Canada
| | - Jolien De Waele
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Jérôme Montnach
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - Sandrine Cestèle
- Université Cote d'Azur, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
| | - Agnès Tessier
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France
| | - J P Johnson
- Xenon Pharmaceuticals, Burnaby, British Columbia, Canada
| | - Massimo Mantegazza
- Université Cote d'Azur, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
| | - Jan Tytgat
- University of Leuven, 3000 Leuven, Belgium
| | - Charles Cohen
- Xenon Pharmaceuticals, Burnaby, British Columbia, Canada
| | | | - Frank Bosmans
- Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium
| | - Céline Landon
- Center for Molecular Biophysics, CNRS, rue Charles Sadron, CS 80054, Orléans 45071, France
| | - Michel De Waard
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000 Nantes, France; Smartox Biotechnology, Saint-Egrève, France; LabEx "Ion Channels, Science and Therapeutics", Valbonne, France.
| |
Collapse
|
38
|
Abstract
In recent years, there has been a significant increase in preclinical studies to test genetic therapies for epilepsy. Some of these therapies have advanced to clinical trials and are being tested in patients with monogenetic or focal refractory epilepsy. This article provides an overview of the current state of preclinical studies that show potential for clinical translation. Specifically, we focus on genetic therapies that have demonstrated a clear effect on seizures in animal models and have the potential to be translated to clinical settings. Both therapies targeting the cause of the disease and those that treat symptoms are discussed. We believe that the next few years will be crucial in determining the potential of genetic therapies for treating patients with epilepsy.
Collapse
Affiliation(s)
- James S. Street
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
39
|
Silva-Cardoso GK, N'Gouemo P. Seizure-suppressor genes: can they help spearhead the discovery of novel therapeutic targets for epilepsy? Expert Opin Ther Targets 2023; 27:657-664. [PMID: 37589085 PMCID: PMC10528013 DOI: 10.1080/14728222.2023.2248375] [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: 04/30/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Epilepsies are disorders of neuronal excitability characterized by spontaneously recurrent focal and generalized seizures, some of which result from genetic mutations. Despite the availability of antiseizure medications, pharmaco-resistant epilepsy is seen in about 23% of epileptic patients worldwide. Therefore, there is an urgent need to develop novel therapeutic strategies for epilepsies. Several epilepsy-associated genes have been found in humans. Seizure susceptibility can also be induced in Drosophila mutants, some showing features resembling human epilepsies. Interestingly, several second-site mutation gene products have been found to suppress seizure susceptibility in the seizure genetic model Drosophila. Thus, these so-called 'seizure-suppressor' gene variants may lead to developing a novel class of antiseizure medications. AREA COVERED This review evaluates the potential therapeutic of seizure-suppressor gene variants. EXPERT OPINION Studies on epilepsy-associated genes have allowed analyses of mutations linked to human epilepsy by reproducing these mutations in Drosophila using reverse genetics to generate potential antiseizure therapeutics. As a result, about fifteen seizure-suppressor gene mutants have been identified. Furthermore, some of these epilepsy gene mutations affect ligand-and voltage-gated ion channels. Therefore, a better understanding of the antiseizure activity of seizure-suppressor genes is essential in advancing gene therapy and precision medicine for epilepsy.
Collapse
Affiliation(s)
- Gleice Kelli Silva-Cardoso
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| | - Prosper N'Gouemo
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC 20059, USA
| |
Collapse
|
40
|
Koch NA, Sonnenberg L, Hedrich UBS, Lauxmann S, Benda J. Loss or gain of function? Effects of ion channel mutations on neuronal firing depend on the neuron type. Front Neurol 2023; 14:1194811. [PMID: 37292138 PMCID: PMC10244640 DOI: 10.3389/fneur.2023.1194811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Clinically relevant mutations to voltage-gated ion channels, called channelopathies, alter ion channel function, properties of ionic currents, and neuronal firing. The effects of ion channel mutations are routinely assessed and characterized as loss of function (LOF) or gain of function (GOF) at the level of ionic currents. However, emerging personalized medicine approaches based on LOF/GOF characterization have limited therapeutic success. Potential reasons are among others that the translation from this binary characterization to neuronal firing is currently not well-understood-especially when considering different neuronal cell types. In this study, we investigate the impact of neuronal cell type on the firing outcome of ion channel mutations. Methods To this end, we simulated a diverse collection of single-compartment, conductance-based neuron models that differed in their composition of ionic currents. We systematically analyzed the effects of changes in ion current properties on firing in different neuronal types. Additionally, we simulated the effects of known mutations in KCNA1 gene encoding the KV1.1 potassium channel subtype associated with episodic ataxia type 1 (EA1). Results These simulations revealed that the outcome of a given change in ion channel properties on neuronal excitability depends on neuron type, i.e., the properties and expression levels of the unaffected ionic currents. Discussion Consequently, neuron-type specific effects are vital to a full understanding of the effects of channelopathies on neuronal excitability and are an important step toward improving the efficacy and precision of personalized medicine approaches.
Collapse
Affiliation(s)
- Nils A. Koch
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| | - Lukas Sonnenberg
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| | - Ulrike B. S. Hedrich
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Stephan Lauxmann
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jan Benda
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, University of Tübingen, Tübingen, Germany
- Bernstein Center for Computational Neuroscience Tübingen, Tübingen, Germany
| |
Collapse
|
41
|
Guo Q, Wu S, Geschwind DH. Characterization of Gene Regulatory Elements in Human Fetal Cortical Development: Enhancing Our Understanding of Neurodevelopmental Disorders and Evolution. Dev Neurosci 2023; 46:69-83. [PMID: 37231806 DOI: 10.1159/000530929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The neocortex is the region that most distinguishes human brain from other mammals and primates [Annu Rev Genet. 2021 Nov;55(1):555-81]. Studying the development of human cortex is important in understanding the evolutionary changes occurring in humans relative to other primates, as well as in elucidating mechanisms underlying neurodevelopmental disorders. Cortical development is a highly regulated process, spatially and temporally coordinated by expression of essential transcriptional factors in response to signaling pathways [Neuron. 2019 Sep;103(6):980-1004]. Enhancers are the most well-understood cis-acting, non-protein-coding regulatory elements that regulate gene expression [Nat Rev Genet. 2014 Apr;15(4):272-86]. Importantly, given the conservation of both DNA sequence and molecular function of the majority of proteins across mammals [Genome Res. 2003 Dec;13(12):2507-18], enhancers [Science. 2015 Mar;347(6226):1155-9], which are far more divergent at the sequence level, likely account for the phenotypes that distinguish the human brain by changing the regulation of gene expression. In this review, we will revisit the conceptual framework of gene regulation during human brain development, as well as the evolution of technologies to study transcriptional regulation, with recent advances in genome biology that open a window allowing us to systematically characterize cis-regulatory elements in developing human brain [Hum Mol Genet. 2022 Oct;31(R1):R84-96]. We provide an update on work to characterize the suite of all enhancers in the developing human brain and the implications for understanding neuropsychiatric disorders. Finally, we discuss emerging therapeutic ideas that utilize our emerging knowledge of enhancer function.
Collapse
Affiliation(s)
- Qiuyu Guo
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
- Center for Autism Research and Treatment, Semel Institute, University of California Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Sarah Wu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
| | - Daniel H Geschwind
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, USA
- Center for Autism Research and Treatment, Semel Institute, University of California Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Institute of Precision Health, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
42
|
Gao C, Pielas M, Jiao F, Mei D, Wang X, Kotulska K, Jozwiak S. Epilepsy in Dravet Syndrome—Current and Future Therapeutic Opportunities. J Clin Med 2023; 12:jcm12072532. [PMID: 37048615 PMCID: PMC10094968 DOI: 10.3390/jcm12072532] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Dravet Syndrome (DS) is a developmental epileptic encephalopathy characterized by drug-resistant seizures and other clinical features, including intellectual disability and behavioral, sleep, and gait problems. The pathogenesis is strongly connected to voltage-gated sodium channel dysfunction. The current consensus of seizure management in DS consists of a combination of conventional and recently approved drugs such as stiripentol, cannabidiol, and fenfluramine. Despite promising results in randomized clinical trials and extension studies, the prognosis of the developmental outcomes of patients with DS remains unfavorable. The article summarizes recent changes in the therapeutic approach to DS and discusses ongoing clinical research directions. Serotonergic agents under investigation show promising results and may replace less DS-specific medicines. The use of antisense nucleotides and gene therapy is focused not only on symptom relief but primarily addresses the underlying cause of the syndrome. Novel compounds, after expected safe and successful implementation in clinical practice, will open a new era for patients with DS. The main goal of causative treatment is to modify the natural course of the disease and provide the best neurodevelopmental outcome with minimum neurological deficit.
Collapse
|
43
|
Toward the Development of Epigenome Editing-Based Therapeutics: Potentials and Challenges. Int J Mol Sci 2023; 24:ijms24054778. [PMID: 36902207 PMCID: PMC10003136 DOI: 10.3390/ijms24054778] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
The advancement in epigenetics research over the past several decades has led to the potential application of epigenome-editing technologies for the treatment of various diseases. In particular, epigenome editing is potentially useful in the treatment of genetic and other related diseases, including rare imprinted diseases, as it can regulate the expression of the epigenome of the target region, and thereby the causative gene, with minimal or no modification of the genomic DNA. Various efforts are underway to successfully apply epigenome editing in vivo, such as improving target specificity, enzymatic activity, and drug delivery for the development of reliable therapeutics. In this review, we introduce the latest findings, summarize the current limitations and future challenges in the practical application of epigenome editing for disease therapy, and introduce important factors to consider, such as chromatin plasticity, for a more effective epigenome editing-based therapy.
Collapse
|
44
|
Westenbroek R, Kaplan J, Viray K, Stella N. The serine hydrolase ABHD6 controls survival and thermally induced seizures in a mouse model of Dravet syndrome. Neurobiol Dis 2023; 180:106099. [PMID: 36990366 DOI: 10.1016/j.nbd.2023.106099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/03/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Evidence suggests that inhibition of α/β hydrolase-domain containing 6 (ABHD6) reduces seizures; however, the molecular mechanism of this therapeutic response remains unknown. We discovered that heterozygous expression of Abhd6 (Abhd6+/-) significantly reduced the premature lethality of Scn1a+/- mouse pups, a genetic mouse model of Dravet Syndrome (DS). Both Abhd6+/- mutation and pharmacological inhibition of ABHD6 reduced the duration and incidence of thermally induced seizures in Scn1a+/- pups. Mechanistically, the in vivo anti-seizure response resulting from ABHD6 inhibition is mediated by potentiation of gamma-aminobutyric acid receptors Type-A (GABAAR). Brain slice electrophysiology showed that blocking ABHD6 potentiates extrasynaptic (tonic) GABAAR currents that reduce dentate granule cell excitatory output without affecting synaptic (phasic) GABAAR currents. Our results unravel an unexpected mechanistic link between ABHD6 activity and extrasynaptic GABAAR currents that controls hippocampal hyperexcitability in a genetic mouse model of DS. BRIEF SUMMARY: This study provides the first evidence for a mechanistic link between ABHD6 activity and the control of extrasynaptic GABAAR currents that controls hippocampal hyperexcitability in a genetic mouse model of Dravet Syndrome and can be targeted to dampened seizures.
Collapse
Affiliation(s)
- Ruth Westenbroek
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Joshua Kaplan
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Psychology, Western Washington University, Bellingham, WA 98225, USA
| | - Katie Viray
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Nephi Stella
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
45
|
Hong D, Iakoucheva LM. Therapeutic strategies for autism: targeting three levels of the central dogma of molecular biology. Transl Psychiatry 2023; 13:58. [PMID: 36792602 PMCID: PMC9931756 DOI: 10.1038/s41398-023-02356-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
The past decade has yielded much success in the identification of risk genes for Autism Spectrum Disorder (ASD), with many studies implicating loss-of-function (LoF) mutations within these genes. Despite this, no significant clinical advances have been made so far in the development of therapeutics for ASD. Given the role of LoF mutations in ASD etiology, many of the therapeutics in development are designed to rescue the haploinsufficient effect of genes at the transcriptional, translational, and protein levels. This review will discuss the various therapeutic techniques being developed from each level of the central dogma with examples including: CRISPR activation (CRISPRa) and gene replacement at the DNA level, antisense oligonucleotides (ASOs) at the mRNA level, and small-molecule drugs at the protein level, followed by a review of current delivery methods for these therapeutics. Since central nervous system (CNS) penetrance is of utmost importance for ASD therapeutics, it is especially necessary to evaluate delivery methods that have higher efficiency in crossing the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Derek Hong
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
46
|
Tan X, Wang J, Yao J, Yuan J, Dai Y, Sun M, Zhang T, Yang J, Cai W, Qiu L, Sun J. Microglia participate in postoperative cognitive dysfunction by mediating the loss of inhibitory synapse through the complement pathway. Neurosci Lett 2023; 796:137049. [PMID: 36608926 DOI: 10.1016/j.neulet.2023.137049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/13/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
BACKGROUND Elderly patients after surgery are prone to cognitive decline known as postoperative cognitive dysfunction (POCD). Several studies have shown that the microglial activation and the increase of complement protein expression in hippocampus induced by surgery may be related to the pathogenesis of POCD. The purpose of this study was to determine whether microglia and complement system were involved in cognitive dysfunction in aged mice. METHODS The POCD model was established by exploratory laparotomy in 15-month-old male C57BL/6J mice and animal behavioral tests were performed to test hippocampal-dependent memory capacity. Minocycline was used to suppress the activation of microglia, and complement 3 receptor inhibitor was used to suppress the association between microglia and complement 3. Western blot and immunofluorescence were used to detect the microglial activation, complement protein, and synaptic protein expressions. RESULTS Operation induced hippocampal-dependent memory impairment (P < 0.01), which was accompanied by microglial activation (P < 0.01). There was also a significant reduction in inhibitory synaptic protein expression in the hippocampus of mice in the surgery group (P < 0.01). However, minocycline, a microglia inhibitor, rescued all the above changes. In addition, C3RI intervention inhibited the phagocytosis of inhibitory synapses by microglia (P < 0.05) and improved the cognitive function of mice (P < 0.01). CONCLUSION Microglia participate in postoperative cognitive dysfunction by mediating inhibitory synaptic loss through the complement pathway.
Collapse
Affiliation(s)
- Xiaoxiang Tan
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Jiajia Wang
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Juan Yao
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Jing Yuan
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Yuchen Dai
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Menghan Sun
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Tianhao Zhang
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Jiaojiao Yang
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Wenlan Cai
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Lili Qiu
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China.
| | - Jie Sun
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
47
|
Abstract
Gene therapy using adeno-associated virus (AAV) is a rapidly developing technology with widespread treatment potential. AAV2 vectors injected directly into the brain by stereotaxic brain surgery have shown good results in treating aromatic l-amino acid decarboxylase deficiency. Moreover, gene therapy using the AAV9 vector, which crosses the blood-brain barrier, has been performed in more than 2000 patients worldwide as a disease-modifying therapy for spinal muscular atrophy. AAV vectors have been applied to the development of gene therapies for various pediatric diseases. Gene therapy trials for hemophilia and ornithine transcarbamylase deficiency are underway. Clinical trials are planned for glucose transporter I deficiency, Niemann-Pick disease type C, and spinocerebellar ataxia type 1. The genome of AAV vectors is located in the episome and is rarely integrated into chromosomes, making the vectors safe. However, serious adverse events such as hepatic failure and thrombotic microangiopathy have been reported, and ongoing studies are focusing on developing more efficient vectors to reduce required dosages.
Collapse
|
48
|
Zhang R, Liu J, Xue G, Yang J, Li D, Tian T, Zhang X, Gao K, Pan Z. Forced activation of dystrophin transcription by CRISPR/dCas9 reduced arrhythmia susceptibility via restoring membrane Nav1.5 distribution. Gene Ther 2023; 30:142-149. [PMID: 35644811 DOI: 10.1038/s41434-022-00348-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 11/09/2022]
Abstract
Dystrophin deficiency due to genetic mutations causes cardiac abnormalities in Duchenne's muscular dystrophy. Dystrophin is also shown to be downregulated in conventional failing hearts. Whether restoration of dystrophin expression possesses any therapeutic potential for conventional heart failure (HF) remains to be examined. HF mouse model was generated by transverse aortic constriction (TAC). In vivo activation of dystrophin transcription was achieved by tail-vein injection of adeno-associated virus 9 carrying CRISPR/dCas system for dystrophin. We found that activation of dystrophin expression in TAC mice significantly reduced the susceptibility to arrhythmia of TAC mice and the mortality rate. We further demonstrated that over-expression of dystrophin increased cardiac conduction of hearts in TAC mice by optical mapping evaluation. Activation of dystrophin expression also increased peak sodium current in isolated ventricular myocytes from hearts of TAC mice as recorded by the patch-clamp technique. Immunoblotting and immunofluorescence showed that increased dystrophin transcription restored the membrane distribution of Nav1.5 in the hearts of TAC mice. In summary, correction of dystrophin downregulation by the CRISPR-dCas9 system reduced the susceptibility to arrhythmia of conventional HF mice through restoring Nav1.5 membrane distribution. This study paved the way to develop a new therapeutic strategy for HF-related ventricular arrhythmia.
Collapse
Affiliation(s)
- Ruixin Zhang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Junwu Liu
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Genlong Xue
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China.,Institute of Heart and Vascular Diseases, Department of Cardiology, and Central Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiming Yang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Desheng Li
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Tao Tian
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiaofang Zhang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Kangyi Gao
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Zhenwei Pan
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Beijing, 2019RU070, China. .,NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
49
|
Myers KA. SCN1A as a therapeutic target for Dravet syndrome. Expert Opin Ther Targets 2023; 27:459-467. [PMID: 37364240 DOI: 10.1080/14728222.2023.2230364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/08/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION Dravet syndrome is a severe early infancy-onset developmental and epileptic encephalopathy. Patients have drug-resistant seizures, as well as significant co-morbidities, including developmental impairment, crouch gait, sleep disturbance, and early mortality. The underlying cause is mutations in SCN1A, encoding the sodium channel subunit NaV1.1, in >90% of patients. At present, approved Dravet syndrome treatments are symptomatic, primarily aimed at reducing seizure frequency, but having little to no effect on co-morbidities. AREAS COVERED We discuss the potential to treat Dravet syndrome by targeting NaV1.1 directly. Anti-seizure medications that act as sodium channel inhibitors are generally minimally effective and can actually exacerbate seizures. However, other interventions are currently under investigation, including gene therapies that increase the amount of functional NaV1.1. Some of these interventions have encouraging pre-clinical data from in vitro and animal models. EXPERT OPINION Increasing functional NaV1.1 via antisense oligonucleotides or virus-borne vectors is the most promising avenue for meaningful improvement in Dravet syndrome treatment, with the potential to not only reduce seizures but also address the multiple co-morbidities associated with this disease. However, human clinical trial data are necessary to determine safety and to clarify if, and to what extent, these interventions modify the natural history of Dravet syndrome.
Collapse
Affiliation(s)
- Kenneth A Myers
- Child Health and Human Development Program, Research Institute of the McGill University Medical Centre, Montreal, Quebec, Canada
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Operto FF, Pastorino GMG, Viggiano A, Dell’Isola GB, Dini G, Verrotti A, Coppola G. Epilepsy and Cognitive Impairment in Childhood and Adolescence: A Mini-Review. Curr Neuropharmacol 2023; 21:1646-1665. [PMID: 35794776 PMCID: PMC10514538 DOI: 10.2174/1570159x20666220706102708] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/28/2022] [Accepted: 04/26/2022] [Indexed: 11/22/2022] Open
Abstract
Managing epilepsy in people with an intellectual disability remains a therapeutic challenge and must take into account additional issues such as diagnostic difficulties and frequent drug resistance. Advances in genomic technologies improved our understanding of epilepsy and raised the possibility to develop patients-tailored treatments acting on the key molecular mechanisms involved in the development of the disease. In addition to conventional antiseizure medications (ASMs), ketogenic diet, hormone therapy and epilepsy surgery play an important role, especially in cases of drugresistance. This review aims to provide a comprehensive overview of the mainfactors influencing cognition in children and adolescents with epilepsy and the main therapeutic options available for the epilepsies associated with intellectual disability.
Collapse
Affiliation(s)
- Francesca Felicia Operto
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Grazia Maria Giovanna Pastorino
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, SA, Italy
| | | | - Gianluca Dini
- Department of Pediatrics, University of Perugia, Giorgio Menghini Square, 06129 Perugia, Italy
| | - Alberto Verrotti
- Department of Pediatrics, University of Perugia, Giorgio Menghini Square, 06129 Perugia, Italy
| | - Giangennaro Coppola
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| |
Collapse
|