1
|
Jain N, Roy AA, Madhusoodanan G, Preman NK, Pokale R, Pisay M, Mukharya A, Pandey A, Mutalik S. Unlocking the future: Precision oligonucleotide therapy for targeted treatment of neurodegenerative disorders. Int J Biol Macromol 2025; 310:143515. [PMID: 40288719 DOI: 10.1016/j.ijbiomac.2025.143515] [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: 03/20/2025] [Revised: 04/09/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Neurodegenerative disorders are complex and devastating conditions of the central nervous system that profoundly impact quality of life. Given the limited treatment options available, there is a pressing need to develop novel therapeutic strategies. Oligonucleotides have emerged as key players in precision medicine for these disorders, but their potential is hindered by poor translocation across the blood-brain barrier. This review focuses on neurodegenerative disorders other than Alzheimer's and Parkinson's, which are widely reported in the literature, and aims to address the significant hurdles in oligonucleotide delivery for neurodegenerative diseases. It highlights recent advancements in CNS-targeting approaches, such as chemical conjugation, antibody-oligonucleotide conjugates, focused ultrasound, and viral and nanocarrier-based delivery systems. Each strategy's strengths and limitations are discussed, with potential solutions proposed for more effective treatments. Additionally, the review offers valuable insights into regulatory requirements and prospects for clinical translation, which are crucial for shaping the future of neurodegenerative therapies. By exploring these innovative approaches, the goal is to surmount challenges posed by the blood-brain barrier and develop more effective treatments, thereby enhancing the quality of life of the patients suffering from these debilitating conditions.
Collapse
Affiliation(s)
- Naitik Jain
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Geethu Madhusoodanan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Namitha K Preman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Rahul Pokale
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Muralidhar Pisay
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Anoushka Mukharya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Abhijeet Pandey
- Novartis Healthcare Private Ltd, Novartis Knowledge Center, Hyderabad 500081, Telangana, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
2
|
Wang Y, Cheng Y, Sun H, Wang Z, Chen N, Shi C, Liu H, Yang J, Xu Y. Genome editing in spinocerebellar ataxia type 3 cells improves Golgi apparatus structure. Sci Rep 2025; 15:12106. [PMID: 40204795 PMCID: PMC11982189 DOI: 10.1038/s41598-025-93369-8] [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: 09/12/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is an autosomal dominant neurodegenerative disease caused by repeat expansion of the CAG trinucleotide within exon 10 of the ATXN3 gene. This mutation results in the production of an abnormal ataxin-3 protein containing an extended polyglutamine tract, referred to as mutant ataxin-3. In this study, we investigated the therapeutic potential of CRISPR/Cas9-mediated genome editing for SCA3. First, we designed a specific single-guide RNA targeting the ATXN3 gene and constructed the corresponding targeting vector. Induced pluripotent stem cells (iPSCs) derived from a SCA3 patient were then electroporated with the CRISPR/Cas9 components. Positive clones were screened and validated by PCR and Sanger sequencing to obtain genome-editing iPSCs (GE-iPSCs). Subsequently, the pluripotency of GE-iPSCs was confirmed, and the effects of genome editing on mutant ataxin-3 protein expression and Golgi apparatus morphology were assessed using Western blotting and immunofluorescence analyses. Our results demonstrated that targeted insertion of polyadenylation signals (PAS) upstream of the abnormal CAG repeats effectively suppressed the production of mutant ataxin-3. This intervention also reduced the formation of neuronal nuclear inclusions in differentiated neurons, restored the structural integrity of the Golgi apparatus (which exhibited a loose and enlarged morphology in SCA3 cells), and increased the expression levels of Golgi structural proteins (GM130 and GORASP2). In conclusion, our findings indicate that the targeted insertion of PAS upstream of the abnormal CAG repeats in the ATXN3 gene represents a promising therapeutic strategy for SCA3 through genome editing.
Collapse
Affiliation(s)
- Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Yunan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Huifang Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Zhuoya Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Na Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China.
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China.
| |
Collapse
|
3
|
Cheng Y, Gai C, Zhao Y, Li T, Song Y, Luo Q, Xin D, Jiang Z, Chen W, Liu D, Wang Z. Engineered Extracellular Vesicles Loaded with MiR-100-5p Antagonist Selectively Target the Lesioned Region to Promote Recovery from Brain Damage. Neurosci Bull 2025:10.1007/s12264-025-01376-6. [PMID: 40167866 DOI: 10.1007/s12264-025-01376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 11/05/2024] [Indexed: 04/02/2025] Open
Abstract
Hypoxic-ischemic (HI) brain damage poses a high risk of death or lifelong disability, yet effective treatments remain elusive. Here, we demonstrated that miR-100-5p levels in the lesioned cortex increased after HI insult in neonatal mice. Knockdown of miR-100-5p expression in the brain attenuated brain injury and promoted functional recovery, through inhibiting the cleaved-caspase-3 level, microglia activation, and the release of proinflammation cytokines following HI injury. Engineered extracellular vesicles (EVs) containing neuron-targeting rabies virus glycoprotein (RVG) and miR-100-5p antagonists (RVG-EVs-Antagomir) selectively targeted brain lesions and reduced miR-100-5p levels after intranasal delivery. Both pre- and post-HI administration showed therapeutic benefits. Mechanistically, we identified protein phosphatase 3 catalytic subunit alpha (Ppp3ca) as a novel candidate target gene of miR-100-5p, inhibiting c-Fos expression and neuronal apoptosis following HI insult. In conclusion, our non-invasive method using engineered EVs to deliver miR-100-5p antagomirs to the brain significantly improves functional recovery after HI injury by targeting Ppp3ca to suppress neuronal apoptosis.
Collapse
Affiliation(s)
- Yahong Cheng
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Danqing Xin
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zige Jiang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wenqiang Chen
- Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
4
|
Li Z, Du X, Yang Y, Zhang L, Chen P, Kan Y, Pan J, Lin L, Liu D, Jiang X, Zhang CY, Pei Z, Chen X. Treatment of neurological pathology and inflammation in Machado-Joseph disease through in vivo self-assembled siRNA. Brain 2025; 148:817-832. [PMID: 39315766 PMCID: PMC11884698 DOI: 10.1093/brain/awae304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/23/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Machado-Joseph disease, also known as spinocerebellar ataxia type 3 (MJD/SCA3), is a fatal autosomal dominant hereditary ataxia characterized by cerebellar ataxia resulting from the abnormal expansion of CAG repeats in exon 10 of the ATXN3 gene. At present, there is no effective treatment for SCA3. Small interfering RNAs (siRNAs) are emerging as potential therapeutic strategies to target the disease-causing mutant ATXN3 (mATXN3) protein specifically. However, the efficiency of delivery of siRNAs remains a major obstacle for clinical application, particularly in brain disorders. The aim of this study was to develop a synthetic biology strategy to reprogram the host liver as a tissue chassis to induce and deliver in vivo self-assembled siRNAs to target the ATXN3 gene. A synthetic construct directed by a cytomegalovirus promoter was designed to encode a neuron-targeting rabies virus glycoprotein tag and mATXN3-siRNA. After intravenous injection, the synthetic construct was taken up by mouse livers, which were then reprogrammed to enable the self-assembly, production and secretion of small extracellular vesicles encapsulating mATXN3-siRNA. The small extracellular vesicle-encapsulated mATXN3-siRNA was transported through the endogenous circulating system of small extracellular vesicles, crossing the blood-brain barrier and reaching the cerebellar cortex and spinal cerebellar tract, where they silenced the ATXN3 gene. Treatment with the synthetic construct for 8 or 12 weeks led to significant improvements in motor balance ability and reduction of cerebellar atrophy in YACMJD84.2 transgenic mice. The number of Purkinje cells in the cerebellar cortex was significantly increased, and the loss of myelin basic protein was reduced. Moreover, the quantity of neurotoxic nuclear inclusion bodies and the expression of glial fibrillary acidic protein, which promotes neuroinflammation in activated astrocytes, were decreased significantly. The synthetic construct facilitated the generation and delivery of in vivo self-assembled siRNA to the cerebellar cortex and spinal cerebellar tract, thereby inhibiting the expression of mATXN3 protein. This treatment successfully addressed motor impairments, alleviated neuropathological phenotypes and mitigated neuroinflammation in YACMJD84.2 transgenic mice. Our strategy effectively overcomes the primary challenges associated with siRNA therapy for cerebellar ataxia, offering a promising avenue for future clinical treatments.
Collapse
Affiliation(s)
- Zhizong Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xinghu Du
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yixuan Yang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Li Zhang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Penglu Chen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yansheng Kan
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Jinmeng Pan
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Lishan Lin
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Ding Liu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xiaohong Jiang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210023, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xi Chen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
5
|
Rodrigues B, Leitão RA, Santos M, Trofimov A, Silva M, Inácio ÂS, Abreu M, Nobre RJ, Costa J, Cardoso AL, Milosevic I, Peça J, Oliveiros B, Pereira de Almeida L, Pinheiro PS, Carvalho AL. MiR-186-5p inhibition restores synaptic transmission and neuronal network activity in a model of chronic stress. Mol Psychiatry 2025; 30:1034-1046. [PMID: 39237722 PMCID: PMC11835755 DOI: 10.1038/s41380-024-02715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Chronic stress exerts profound negative effects on cognitive and emotional behaviours and is a major risk factor for the development of neuropsychiatric disorders. However, the molecular links between chronic stress and its deleterious effects on neuronal and synaptic function remain elusive. Here, using a combination of in vitro and in vivo approaches, we demonstrate that the upregulation of miR-186-5p triggered by chronic stress may be a key mediator of such changes, leading to synaptic dysfunction. Our results show that the expression levels of miR-186-5p are increased both in the prefrontal cortex (PFC) of mice exposed to chronic stress and in cortical neurons chronically exposed to dexamethasone. Additionally, viral overexpression of miR-186-5p in the PFC of naïve mice induces anxiety- and depressive-like behaviours. The upregulation of miR-186-5p through prolonged glucocorticoid receptor activation in vitro, or in a mouse model of chronic stress, differentially affects glutamatergic and GABAergic synaptic transmission, causing an imbalance in excitation/inhibition that leads to altered neuronal network activity. At glutamatergic synapses, we observed both a reduction in synaptic AMPARs and synaptic transmission, whereas GABAergic synaptic transmission was strengthened. These changes could be rescued in vitro by a miR-186-5p inhibitor. Overall, our results establish a novel molecular link between chronic glucocorticoid receptor activation, the upregulation of miR-186-5p and the synaptic changes induced by chronic stress, that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Beatriz Rodrigues
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ricardo A Leitão
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Santos
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Alexander Trofimov
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Integrative Brain Function Neurobiology Lab, I.P. Pavlov Department of Physiology, Institute of Experimental Medicine, 197022, St. Petersburg, Russia
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 010000, Astana, Kazakhstan
| | - Mariline Silva
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Department of Applied Physics and Science for Life Laboratory (SciLifeLab), KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Ângela S Inácio
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Mónica Abreu
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rui J Nobre
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jéssica Costa
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- Experimental Biology and Biomedicine Doctoral Programme, Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ana Luísa Cardoso
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Ira Milosevic
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Multidisciplinary Institute of Aging, MIA Portugal, University of Coimbra, 3004-504, Coimbra, Portugal
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - João Peça
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal
| | - Bárbara Oliveiros
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- iCRB-Coimbra Institute for Clinical and Biomedical Research, University of Coimbra, 3000-548, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- ViraVector, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Paulo S Pinheiro
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- CiBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456, Coimbra, Portugal.
| |
Collapse
|
6
|
Diez-Roda P, Perez-Navarro E, Garcia-Martin R. Adipose Tissue as a Major Launch Spot for Circulating Extracellular Vesicle-Carried MicroRNAs Coordinating Tissue and Systemic Metabolism. Int J Mol Sci 2024; 25:13488. [PMID: 39769251 PMCID: PMC11677924 DOI: 10.3390/ijms252413488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Circulating microRNAs (miRNAs), especially transported by extracellular vesicles (EVs), have recently emerged as major new participants in interorgan communication, playing an important role in the metabolic coordination of our tissues. Among these, adipose tissue displays an extraordinary ability to secrete a vast list of EV-carried miRNAs into the circulation, representing new hormone-like factors. Despite the limitations of current methodologies for the unequivocal identification of the origin and destination of EV-carried miRNAs in vivo, recent investigations clearly support the important regulatory role of adipose-derived circulating miRNAs in shaping the metabolism and function of other tissues including the liver, muscle, endocrine pancreas, cardiovascular system, gastrointestinal tract, and brain. Here, we review the most recent findings regarding miRNAs transported by adipose-derived EVs (AdEVs) targeting other major metabolic organs and the implications of this dialog for physiology and pathology. We also review here the current and potential future diagnostic and therapeutic applications of AdEV-carried miRNAs.
Collapse
Affiliation(s)
| | | | - Ruben Garcia-Martin
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, 28049 Madrid, Spain; (P.D.-R.); (E.P.-N.)
| |
Collapse
|
7
|
Zhao S, Di Y, Fan H, Xu C, Li H, Wang Y, Wang W, Li C, Wang J. Targeted delivery of extracellular vesicles: the mechanisms, techniques and therapeutic applications. MOLECULAR BIOMEDICINE 2024; 5:60. [PMID: 39567444 PMCID: PMC11579273 DOI: 10.1186/s43556-024-00230-x] [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: 08/14/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-derived vesicles with a phospholipid bilayer measuring 50-150 nm in diameter with demonstrated therapeutic potentials. Limitations such as the natural biodistribution (mainly concentrated in the liver and spleen) and short plasma half-life of EVs present significant challenges to their clinical translation. In recent years, growing research indicated that engineered EVs with enhanced targeting to lesion sites have markedly promoted therapeutic efficacy. However, there is a dearth of systematic knowledge on the recent advances in engineering EVs for targeted delivery. Herein, we provide an overview of the targeting mechanisms, engineering techniques, and clinical translations of natural and engineered EVs in therapeutic applications. Enrichment of EVs at lesion sites may be achieved through the recognition of tissue markers, pathological changes, and the circumvention of mononuclear phagocyte system (MPS). Alternatively, external stimuli, including magnetic fields and ultrasound, may also be employed. EV engineering techniques that fulfill targeting functions includes genetic engineering, membrane fusion, chemical modification and physical modification. A comparative statistical analysis was conducted to elucidate the discrepancies between the diverse techniques on size, morphology, stability, targeting and therapeutic efficacy in vitro and in vivo. Additionally, a summary of the registered clinical trials utilizing EVs from 2010 to 2023 has been provided, with a full discussion on the perspectives. This review provides a comprehensive overview of the mechanisms and techniques associated with targeted delivery of EVs in therapeutic applications to advocate further explorations of engineered EVs and accelerate their clinical applications.
Collapse
Affiliation(s)
- Shuang Zhao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yunfeng Di
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huilan Fan
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chengyan Xu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haijing Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
| | - Wei Wang
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chun Li
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyu Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
8
|
Zenge C, Ordureau A. Ubiquitin system mutations in neurological diseases. Trends Biochem Sci 2024; 49:875-887. [PMID: 38972780 PMCID: PMC11455613 DOI: 10.1016/j.tibs.2024.06.011] [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: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Neuronal ubiquitin balance impacts the fate of countless cellular proteins, and its disruption is associated with various neurological disorders. The ubiquitin system is critical for proper neuronal cell state transitions and the clearance of misfolded or aggregated proteins that threaten cellular integrity. This article reviews the state of and recent advancements in our understanding of the disruptions to components of the ubiquitin system, in particular E3 ligases and deubiquitylases, in neurodevelopmental and neurodegenerative diseases. Specific focus is on enzymes with recent progress in their characterization, including identifying enzyme-substrate pairs, the use of stem cell and animal models, and the development of therapeutics for ubiquitin-related diseases.
Collapse
Affiliation(s)
- Colin Zenge
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
9
|
Brezgin S, Danilik O, Yudaeva A, Kachanov A, Kostyusheva A, Karandashov I, Ponomareva N, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. Basic Guide for Approaching Drug Delivery with Extracellular Vesicles. Int J Mol Sci 2024; 25:10401. [PMID: 39408730 PMCID: PMC11476574 DOI: 10.3390/ijms251910401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Extracellular vesicles (EVs) are natural carriers of biomolecules that play a crucial role in cell-to-cell communication and tissue homeostasis under normal and pathological conditions, including inflammatory diseases and cancer. Since the discovery of the pro-regenerative and immune-modulating properties of EVs, EV-based therapeutics have entered clinical trials for conditions such as myocardial infarction and autoimmune diseases, among others. Due to their unique advantages-such as superior bioavailability, substantial packaging capacity, and the ability to traverse biological barriers-EVs are regarded as a promising platform for targeted drug delivery. However, achieving a sufficient accumulation of therapeutic agents at the target site necessitates a larger quantity of EVs per dose compared to using EVs as standalone drugs. This challenge can be addressed by administering larger doses of EVs, increasing the drug dosage per administration, or enhancing the selective accumulation of EVs at target cells. In this review, we will discuss methods to improve the isolation and purification of EVs, approaches to enhance cargo packaging-including proteins, RNAs, and small-molecule drugs-and technologies for displaying targeting ligands on the surface of EVs to facilitate improved targeting. Ultimately, this guide can be applied to the development of novel classes of EV-based therapeutics and to overcoming existing technological challenges.
Collapse
Affiliation(s)
- Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Oleg Danilik
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
| | - Ivan Karandashov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (S.B.); (A.Y.); (A.K.); (A.K.); (I.K.); (N.P.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
10
|
de Sousa-Lourenço J, Silva AC, Pereira de Almeida L, Nobre RJ. Molecular therapy for polyQ disorders: from bench to clinical trials. Trends Mol Med 2024; 30:804-808. [PMID: 38839514 DOI: 10.1016/j.molmed.2024.05.004] [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: 03/18/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Polyglutamine (polyQ) disorders are monogenic neurodegenerative disorders. Currently, no therapies are available for this complex group of disorders. Here, we aim to provide an overview of recent promising preclinical studies and the ongoing clinical trials focusing on molecular therapies for polyQ disorders.
Collapse
Affiliation(s)
- João de Sousa-Lourenço
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana C Silva
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal; GeneT, Gene Therapy Center of Excellence, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; GeneT, Gene Therapy Center of Excellence, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; ViraVector, Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal.
| | - Rui J Nobre
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; GeneT, Gene Therapy Center of Excellence, University of Coimbra, Coimbra, Portugal; ViraVector, Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
11
|
Leandro K, Rufino-Ramos D, Breyne K, Di Ianni E, Lopes SM, Jorge Nobre R, Kleinstiver BP, Perdigão PRL, Breakefield XO, Pereira de Almeida L. Exploring the potential of cell-derived vesicles for transient delivery of gene editing payloads. Adv Drug Deliv Rev 2024; 211:115346. [PMID: 38849005 PMCID: PMC11366383 DOI: 10.1016/j.addr.2024.115346] [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: 12/10/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Gene editing technologies have the potential to correct genetic disorders by modifying, inserting, or deleting specific DNA sequences or genes, paving the way for a new class of genetic therapies. While gene editing tools continue to be improved to increase their precision and efficiency, the limited efficacy of in vivo delivery remains a major hurdle for clinical use. An ideal delivery vehicle should be able to target a sufficient number of diseased cells in a transient time window to maximize on-target editing and mitigate off-target events and immunogenicity. Here, we review major advances in novel delivery platforms based on cell-derived vesicles - extracellular vesicles and virus-like particles - for transient delivery of gene editing payloads. We discuss major findings regarding packaging, in vivo biodistribution, therapeutic efficacy, and safety concerns of cell-derived vesicles delivery of gene editing cargos and their potential for clinical translation.
Collapse
Affiliation(s)
- Kevin Leandro
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal
| | - David Rufino-Ramos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal; Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Koen Breyne
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02129, USA
| | - Emilio Di Ianni
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02129, USA
| | - Sara M Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; ViraVector - Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra 3004-504, Portugal
| | - Benjamin P Kleinstiver
- Center for Genomic Medicine and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Pedro R L Perdigão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Xandra O Breakefield
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA 02129, USA
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; GeneT - Gene Therapy Center of Excellence Portugal, University of Coimbra, Coimbra, Portugal; ViraVector - Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
12
|
Henriques C, Lopes MM, Silva AC, Lobo DD, Badin RA, Hantraye P, Pereira de Almeida L, Nobre RJ. Viral-based animal models in polyglutamine disorders. Brain 2024; 147:1166-1189. [PMID: 38284949 DOI: 10.1093/brain/awae012] [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: 07/09/2023] [Revised: 11/26/2023] [Accepted: 12/30/2023] [Indexed: 01/30/2024] Open
Abstract
Polyglutamine disorders are a complex group of incurable neurodegenerative disorders caused by an abnormal expansion in the trinucleotide cytosine-adenine-guanine tract of the affected gene. To better understand these disorders, our dependence on animal models persists, primarily relying on transgenic models. In an effort to complement and deepen our knowledge, researchers have also developed animal models of polyglutamine disorders employing viral vectors. Viral vectors have been extensively used to deliver genes to the brain, not only for therapeutic purposes but also for the development of animal models, given their remarkable flexibility. In a time- and cost-effective manner, it is possible to use different transgenes, at varying doses, in diverse targeted tissues, at different ages, and in different species, to recreate polyglutamine pathology. This paper aims to showcase the utility of viral vectors in disease modelling, share essential considerations for developing animal models with viral vectors, and provide a comprehensive review of existing viral-based animal models for polyglutamine disorders.
Collapse
Affiliation(s)
- Carina Henriques
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel M Lopes
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana C Silva
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Diana D Lobo
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Romina Aron Badin
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Philippe Hantraye
- CEA, DRF, Institute of Biology François Jacob, Molecular Imaging Research Center (MIRCen), 92265 Fontenay-aux-Roses, France
- CNRS, CEA, Paris-Sud University, Université Paris-Saclay, Neurodegenerative Diseases Laboratory (UMR9199), 92265 Fontenay-aux-Roses, France
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Jorge Nobre
- Center for Neuroscience and Cell Biology (CNC), Gene and Stem Cell Therapies for the Brain Group, University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
- ViraVector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research (III), University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
13
|
Erana-Perez Z, Igartua M, Santos-Vizcaino E, Hernandez RM. Genetically engineered loaded extracellular vesicles for drug delivery. Trends Pharmacol Sci 2024; 45:350-365. [PMID: 38508958 DOI: 10.1016/j.tips.2024.02.006] [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: 12/22/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024]
Abstract
The use of extracellular vesicles (EVs) for drug delivery is being widely explored by scientists from several research fields. To fully exploit their therapeutic potential, multiple methods for loading EVs have been developed. Although exogenous methods have been extensively utilized, in recent years the endogenous method has gained significant attention. This approach, based on parental cell genetic engineering, is suitable for loading large therapeutic biomolecules such as proteins and nucleic acids. We review the most commonly used EV loading methods and emphasize the inherent advantages of the endogenous method over the others. We also examine the most recent advances and applications of this innovative approach to inform on the diverse therapeutic opportunities that lie ahead in the field of EV-based therapies.
Collapse
Affiliation(s)
- Zuriñe Erana-Perez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain.
| |
Collapse
|
14
|
Rhim WK, Kim JY, Lee SY, Cha SG, Park JM, Park HJ, Park CG, Han DK. Recent advances in extracellular vesicle engineering and its applications to regenerative medicine. Biomater Res 2023; 27:130. [PMID: 38082304 PMCID: PMC10712135 DOI: 10.1186/s40824-023-00468-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 11/24/2023] [Indexed: 01/02/2025] Open
Abstract
Extracellular vesicles (EVs) are nanosized particles that are released from cells and reflect the characteristics of the mother cell. Recently, the EVs have been used in several types of studies across many different fields. In the field of EV research, multiple cell culture and EV isolation techniques have been highlighted in importance. Various strategies, including exclusive component culture media, three-dimensional (3D) cultures, and hypoxic conditions, have been proposed for the cell culture to control function of the EVs. Ultracentrifugation, ultrafiltration, precipitation, and tangential flow filtration (TFF) have been utilized for EV isolation. Although isolated EVs have their own functionalities, several researchers are trying to functionalize EVs by applying various engineering approaches. Gene editing, exogenous, endogenous, and hybridization methods are the four well-known types of EV functionalization strategies. EV engineered through these processes has been applied in the field of regenerative medicine, including kidney diseases, osteoarthritis, rheumatoid arthritis, nervous system-related diseases, and others. In this review, it was focused on engineering approaches for EV functionalization and their applications in regenerative medicine.
Collapse
Affiliation(s)
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
- Department of Biomedical Engineering, 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Seung Yeon Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jeong Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyeon Jeong Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
15
|
Rufino-Ramos D, Leandro K, Perdigão PRL, O'Brien K, Pinto MM, Santana MM, van Solinge TS, Mahjoum S, Breakefield XO, Breyne K, Pereira de Almeida L. Extracellular communication between brain cells through functional transfer of Cre mRNA mediated by extracellular vesicles. Mol Ther 2023; 31:2220-2239. [PMID: 37194237 PMCID: PMC10362460 DOI: 10.1016/j.ymthe.2023.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023] Open
Abstract
In the central nervous system (CNS), the crosstalk between neural cells is mediated by extracellular mechanisms, including brain-derived extracellular vesicles (bdEVs). To study endogenous communication across the brain and periphery, we explored Cre-mediated DNA recombination to permanently record the functional uptake of bdEVs cargo over time. To elucidate functional cargo transfer within the brain at physiological levels, we promoted the continuous secretion of physiological levels of neural bdEVs containing Cre mRNA from a localized region in the brain by in situ lentiviral transduction of the striatum of Flox-tdTomato Ai9 mice reporter of Cre activity. Our approach efficiently detected in vivo transfer of functional events mediated by physiological levels of endogenous bdEVs throughout the brain. Remarkably, a spatial gradient of persistent tdTomato expression was observed along the whole brain, exhibiting an increment of more than 10-fold over 4 months. Moreover, bdEVs containing Cre mRNA were detected in the bloodstream and extracted from brain tissue to further confirm their functional delivery of Cre mRNA in a novel and highly sensitive Nanoluc reporter system. Overall, we report a sensitive method to track bdEV transfer at physiological levels, which will shed light on the role of bdEVs in neural communication within the brain and beyond.
Collapse
Affiliation(s)
- David Rufino-Ramos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Kevin Leandro
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Pedro R L Perdigão
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Killian O'Brien
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Maria Manuel Pinto
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Magda M Santana
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Thomas S van Solinge
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Shadi Mahjoum
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Xandra O Breakefield
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Koen Breyne
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|