1
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2024; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
2
|
Ebrahimi P, Davoudi E, Sadeghian R, Zadeh AZ, Razmi E, Heidari R, Morowvat MH, Sadeghian I. In vivo and ex vivo gene therapy for neurodegenerative diseases: a promise for disease modification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7501-7530. [PMID: 38775852 DOI: 10.1007/s00210-024-03141-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/01/2024] [Indexed: 10/04/2024]
Abstract
Neurodegenerative diseases (NDDs), including AD, PD, HD, and ALS, represent a growing public health concern linked to aging and lifestyle factors, characterized by progressive nervous system damage leading to motor and cognitive deficits. Current therapeutics offer only symptomatic management, highlighting the urgent need for disease-modifying treatments. Gene therapy has emerged as a promising approach, targeting the underlying pathology of diseases with diverse strategies including gene replacement, gene silencing, and gene editing. This innovative therapeutic approach involves introducing functional genetic material to combat disease mechanisms, potentially offering long-term efficacy and disease modification. With advancements in genomics, structural biology, and gene editing tools such as CRISPR/Cas9, gene therapy holds significant promise for addressing the root causes of NDDs. Significant progress in preclinical and clinical studies has demonstrated the potential of in vivo and ex vivo gene therapy to treat various NDDs, offering a versatile and precise approach in comparison to conventional treatments. The current review describes various gene therapy approaches employed in preclinical and clinical studies for the treatment of NDDs, including AD, PD, HD, and ALS, and addresses some of the key translational challenges in this therapeutic approach.
Collapse
Affiliation(s)
- Pouya Ebrahimi
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Davoudi
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | | | - Amin Zaki Zadeh
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Emran Razmi
- Arak University of Medical Sciences, Arak, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Morowvat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Quan M, Cao S, Wang Q, Wang S, Jia J. Genetic Phenotypes of Alzheimer's Disease: Mechanisms and Potential Therapy. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:333-349. [PMID: 37589021 PMCID: PMC10425323 DOI: 10.1007/s43657-023-00098-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 08/18/2023]
Abstract
Years of intensive research has brought us extensive knowledge on the genetic and molecular factors involved in Alzheimer's disease (AD). In addition to the mutations in the three main causative genes of familial AD (FAD) including presenilins and amyloid precursor protein genes, studies have identified several genes as the most plausible genes for the onset and progression of FAD, such as triggering receptor expressed on myeloid cells 2, sortilin-related receptor 1, and adenosine triphosphate-binding cassette transporter subfamily A member 7. The apolipoprotein E ε4 allele is reported to be the strongest genetic risk factor for sporadic AD (SAD), and it also plays an important role in FAD. Here, we reviewed recent developments in genetic and molecular studies that contributed to the understanding of the genetic phenotypes of FAD and compared them with SAD. We further reviewed the advancements in AD gene therapy and discussed the future perspectives based on the genetic phenotypes.
Collapse
Affiliation(s)
- Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
| | - Shuman Cao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
| | - Shiyuan Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053 China
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053 China
- Center of Alzheimer’s Disease, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100053 China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053 China
| |
Collapse
|
4
|
Bharti S, Anant PS, Kumar A. Nanotechnology in stem cell research and therapy. JOURNAL OF NANOPARTICLE RESEARCH 2023; 25:6. [DOI: 10.1007/s11051-022-05654-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
|
5
|
iPSCs in Neurodegenerative Disorders: A Unique Platform for Clinical Research and Personalized Medicine. J Pers Med 2022; 12:jpm12091485. [PMID: 36143270 PMCID: PMC9500601 DOI: 10.3390/jpm12091485] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022] Open
Abstract
In the past, several animal disease models were developed to study the molecular mechanism of neurological diseases and discover new therapies, but the lack of equivalent animal models has minimized the success rate. A number of critical issues remain unresolved, such as high costs for developing animal models, ethical issues, and lack of resemblance with human disease. Due to poor initial screening and assessment of the molecules, more than 90% of drugs fail during the final step of the human clinical trial. To overcome these limitations, a new approach has been developed based on induced pluripotent stem cells (iPSCs). The discovery of iPSCs has provided a new roadmap for clinical translation research and regeneration therapy. In this article, we discuss the potential role of patient-derived iPSCs in neurological diseases and their contribution to scientific and clinical research for developing disease models and for developing a roadmap for future medicine. The contribution of humaniPSCs in the most common neurodegenerative diseases (e.g., Parkinson’s disease and Alzheimer’s disease, diabetic neuropathy, stroke, and spinal cord injury) were examined and ranked as per their published literature on PUBMED. We have observed that Parkinson’s disease scored highest, followed by Alzheimer’s disease. Furthermore, we also explored recent advancements in the field of personalized medicine, such as the patient-on-a-chip concept, where iPSCs can be grown on 3D matrices inside microfluidic devices to create an in vitro disease model for personalized medicine.
Collapse
|
6
|
Applications of Ultrasound-Mediated Gene Delivery in Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9050190. [PMID: 35621468 PMCID: PMC9137703 DOI: 10.3390/bioengineering9050190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 11/21/2022] Open
Abstract
Research on the capability of non-viral gene delivery systems to induce tissue regeneration is a continued effort as the current use of viral vectors can present with significant limitations. Despite initially showing lower gene transfection and gene expression efficiencies, non-viral delivery methods continue to be optimized to match that of their viral counterparts. Ultrasound-mediated gene transfer, referred to as sonoporation, occurs by the induction of transient membrane permeabilization and has been found to significantly increase the uptake and expression of DNA in cells across many organ systems. In addition, it offers a more favorable safety profile compared to other non-viral delivery methods. Studies have shown that microbubble-enhanced sonoporation can elicit significant tissue regeneration in both ectopic and disease models, including bone and vascular tissue regeneration. Despite this, no clinical trials on the use of sonoporation for tissue regeneration have been conducted, although current clinical trials using sonoporation for other indications suggest that the method is safe for use in the clinical setting. In this review, we describe the pre-clinical studies conducted thus far on the use of sonoporation for tissue regeneration. Further, the various techniques used to increase the effectiveness and duration of sonoporation-induced gene transfer, as well as the obstacles that may be currently hindering clinical translation, are explored.
Collapse
|
7
|
Parambi DGT, Alharbi KS, Kumar R, Harilal S, Batiha GES, Cruz-Martins N, Magdy O, Musa A, Panda DS, Mathew B. Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:191-233. [PMID: 34655056 PMCID: PMC8518903 DOI: 10.1007/s12035-021-02555-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022]
Abstract
The etiology of many neurological diseases affecting the central nervous system (CNS) is unknown and still needs more effective and specific therapeutic approaches. Gene therapy has a promising future in treating neurodegenerative disorders by correcting the genetic defects or by therapeutic protein delivery and is now an attraction for neurologists to treat brain disorders, like Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, spinocerebellar ataxia, epilepsy, Huntington's disease, stroke, and spinal cord injury. Gene therapy allows the transgene induction, with a unique expression in cells' substrate. This article mainly focuses on the delivering modes of genetic materials in the CNS, which includes viral and non-viral vectors and their application in gene therapy. Despite the many clinical trials conducted so far, data have shown disappointing outcomes. The efforts done to improve outcomes, efficacy, and safety in the identification of targets in various neurological disorders are also discussed here. Adapting gene therapy as a new therapeutic approach for treating neurological disorders seems to be promising, with early detection and delivery of therapy before the neuron is lost, helping a lot the development of new therapeutic options to translate to the clinic.
Collapse
Affiliation(s)
- Della Grace Thomas Parambi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Khalid Saad Alharbi
- College of Pharmacy, Department of Pharmaceutical Chemistry, Jouf University, Al Jouf-2014, Sakaka, Saudi Arabia
| | - Rajesh Kumar
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Seetha Harilal
- Kerala University of Health Sciences, Thrissur, Kerala 680596 India
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Omnia Magdy
- Department of Clinical Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014 Kingdom of Saudi Arabia
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
| | - Arafa Musa
- Pharmacognosy Department, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341 Kingdom of Saudi Arabia
- Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371 Egypt
| | - Dibya Sundar Panda
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Al Jouf, Sakaka, 72341 Kingdom of Saudi Arabia
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041 India
| |
Collapse
|
8
|
Farzamfar S, Nazeri N, Salehi M, Valizadeh A, Marashi SM, Savari Kouzehkonan G, Ghanbari H. Will Nanotechnology Bring New Hope for Stem Cell Therapy? Cells Tissues Organs 2019; 206:229-241. [PMID: 31288229 DOI: 10.1159/000500517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/21/2019] [Indexed: 01/05/2025] Open
Abstract
The potential of stem cell therapy has been shown in preclinical trials for the treatment of damage and replacement of organs and degenerative diseases. After many years of research, its clinical application is limited. Currently there is not a single stem cell therapy product or procedure. Nanotechnology is an emerging field in medicine and has huge potential due to its unique characteristics such as its size, surface effects, tunnel effects, and quantum size effect. The importance of application of nanotechnology in stem cell technology and cell-based therapies has been recognized. In particular, the effects of nanotopography on stem cell differentiation, proliferation, and adhesion have become an area of intense research in tissue engineering and regenerative medicine. Despite the many opportunities that nanotechnology can create to change the fate of stem cell technology and cell therapies, it poses several risks since some nanomaterials are cytotoxic and can affect the differentiation program of stem cells and their viability. Here we review some of the advances and the prospects of nanotechnology in stem cell research and cell-based therapies and discuss the issues, obstacles, applications, and approaches with the aim of opening new avenues for further research.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Nazeri
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Majid Salehi
- Tissue Engineering and Stem Cell Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Alireza Valizadeh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Savari Kouzehkonan
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Islam MA, Firdous J, Badruddoza AZM, Reesor E, Azad M, Hasan A, Lim M, Cao W, Guillemette S, Cho CS. M cell targeting engineered biomaterials for effective vaccination. Biomaterials 2018; 192:75-94. [PMID: 30439573 DOI: 10.1016/j.biomaterials.2018.10.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/09/2018] [Accepted: 10/28/2018] [Indexed: 02/08/2023]
Abstract
Vaccines are one of the greatest medical interventions of all time and have been successful in controlling and eliminating a myriad of diseases over the past two centuries. Among several vaccination strategies, mucosal vaccines have wide clinical applications and attract considerable interest in research, showing potential as innovative and novel therapeutics. In mucosal vaccination, targeting (microfold) M cells is a frontline prerequisite for inducing effective antigen-specific immunostimulatory effects. In this review, we primarily focus on materials engineered for use as vaccine delivery platforms to target M cells. We also describe potential M cell targeting areas, methods to overcome current challenges and limitations of the field. Furthermore, we present the potential of biomaterials engineering as well as various natural and synthetic delivery technologies to overcome the challenges of M cell targeting, all of which are absent in current literature. Finally, we briefly discuss manufacturing and regulatory processes to bring a robust perspective on the feasibility and potential of this next-generation vaccine technology.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Jannatul Firdous
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abu Zayed Md Badruddoza
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emma Reesor
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Mohammad Azad
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Michael Lim
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Wuji Cao
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Simon Guillemette
- Department of Nanotechnology Engineering, University of Waterloo, Waterloo, Canada
| | - Chong Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
10
|
Li YQ, Song FH, Zhong K, Yu GY, Zilundu PLM, Zhou YY, Fu R, Tang Y, Ling ZM, Xu X, Zhou LH. Pre-Injection of Small Interfering RNA (siRNA) Promotes c-Jun Gene Silencing and Decreases the Survival Rate of Axotomy-Injured Spinal Motoneurons in Adult Mice. J Mol Neurosci 2018; 65:400-410. [PMID: 29992498 DOI: 10.1007/s12031-018-1098-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022]
Abstract
Brachial plexus injury is a common clinical peripheral nerve trauma. A series of genes in motoneurons were activated in the corresponding segments of the spinal cord after brachial plexus roots axotomy. The spatial and temporal expression of these genes directly affects the speed of motoneuron axon regeneration and precise target organ reinnervation. In a previous study, we observed the overexpression of c-Jun in motoneurons of the spinal cord ventral horn after brachial plexus injury in rats. However, the relevance of c-Jun expression with respect to the fate of axotomy-induced branchial plexus injury in adult mice remains unknown. In the present study, we explored the function of c-Jun in motoneuron recovery after axotomy. We pre-injected small interfering RNA (siRNA) to knockdown c-Jun expression in mice and examined the effects of the overexpression of c-Jun in motoneurons after the axotomy of the brachial plexus in vivo. Axotomy induced c-Jun overexpression in the ventral horn motoneurons of adult mice from 3 to 14 days after injury. In addition, the pre-injection of siRNA transiently inhibited c-Jun expression and decreased the survival rate of axotomy-injured motoneurons. These findings indicate that the axotomy-induced overexpression of c-Jun plays an important role in the survival of ventral horn motoneurons in adult mice. In addition, the pre-injection of c-Jun siRNA through the brachial plexus stem effectively adjusts c-Jun gene expression at the ipsilateral side.
Collapse
Affiliation(s)
- Ying-Qin Li
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, No.52 Mei Hua East Road, Zhuhai, 519000, Guangdong Province, People's Republic of China
| | - Fa-Huan Song
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | - Ke Zhong
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Guang-Yin Yu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Prince Last Mudenda Zilundu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Ying-Ying Zhou
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Rao Fu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | - Ying Tang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | - Ze-Min Ling
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | - Xiaoying Xu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China
| | - Li-Hua Zhou
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
11
|
Chen L, Long D, Huang S, Yang Q, Hao J, Wu N, Peng L. Evaluation of a novel poly(amidoamine) with pendant aminobutyl group on the cellular properties of transfected bone marrow mesenchymal stem cells. J Biomed Mater Res A 2017; 106:686-697. [PMID: 28986940 DOI: 10.1002/jbm.a.36264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/07/2017] [Accepted: 07/28/2017] [Indexed: 02/05/2023]
Abstract
Stem cell-based gene therapy has been considered in the treatment of many degenerative diseases. Gene-modified stem cells should maintain its reproductive activity without losing stem cell properties, including genetic phenotype and differentiation potential. In the study, a novel poly (amidoamine) with pendant aminobutyl group (PAA-BA) designed by our group was used in the transfection of bone marrow mesenchymal stromal cells (BMSCs) and the cellular properties post-transfection were evaluated, including DNA content, colony forming capacity, genetic phenotype, and multi-directional differentiation. Two classical non-viral gene delivery vectors, polyethylenimine (PEI) and Lipofectamine 2000 (LP2000) were also used. Compared to non-transfected group, PAA-BA showed minor decreased DNA content but maintained BMSCs' phenotype, reproductive activity and multi-differentiation potential (osteogenic, chondrogenic, adipogenic, and neurogenic differentiation). Both PAA-BA and PEI transfected BMSCs demonstrated improved osteogenic differentiation ability at late stage but suppressed adipogenic as well as mature neural differentiation in vitro. LP2000 and PEI transfected BMSCs displayed significantly lower DNA content and reproductive activity. These findings suggest that PAA-BA is one of safe gene delivery vectors in BMSCs transfection and plays a role in stem cell's osteogenic and neurogenic differentiation. This study proposes the potential application of PAA-BA in BMSCs based gene therapy, in particular bone and nerve relative diseases. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 686-697, 2018.
Collapse
Affiliation(s)
- Lili Chen
- Department of Orthopedic Surgery, West China Hospital, Sichuan University; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China.,Division of Health Quarantine, Shenzhen Entry-Exit Inspection and Quarantine Bureau, 518045, China
| | - Dan Long
- Department of Orthopedic Surgery, West China Hospital, Sichuan University; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shishu Huang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qian Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jin Hao
- Program in Biological Sciences in Dental Medicine, Harvard School of Dental Medicine, Boston, Massachusetts, 02115
| | - Nan Wu
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100032, China
| | - Lin Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Gowing G, Svendsen S, Svendsen CN. Ex vivo gene therapy for the treatment of neurological disorders. PROGRESS IN BRAIN RESEARCH 2017; 230:99-132. [PMID: 28552237 DOI: 10.1016/bs.pbr.2016.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ex vivo gene therapy involves the genetic modification of cells outside of the body to produce therapeutic factors and their subsequent transplantation back into patients. Various cell types can be genetically engineered. However, with the explosion in stem cell technologies, neural stem/progenitor cells and mesenchymal stem cells are most often used. The synergy between the effect of the new cell and the additional engineered properties can often provide significant benefits to neurodegenerative changes in the brain. In this review, we cover both preclinical animal studies and clinical human trials that have used ex vivo gene therapy to treat neurological disorders with a focus on Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, and stroke. We highlight some of the major advances in this field including new autologous sources of pluripotent stem cells, safer ways to introduce therapeutic transgenes, and various methods of gene regulation. We also address some of the remaining hurdles including tunable gene regulation, in vivo cell tracking, and rigorous experimental design. Overall, given the current outcomes from researchers and clinical trials, along with exciting new developments in ex vivo gene and cell therapy, we anticipate that successful treatments for neurological diseases will arise in the near future.
Collapse
Affiliation(s)
- Genevieve Gowing
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Soshana Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
13
|
Lin MY, Wu YC, Lee JA, Tung KW, Zhou J, Teitell MA, Yeh JA, Chiou PY. Intracellular Delivery by Shape Anisotropic Magnetic Particle-Induced Cell Membrane Cuts. ACTA ACUST UNITED AC 2016; 21:548-56. [PMID: 26882924 DOI: 10.1177/2211068216630743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Indexed: 11/15/2022]
Abstract
Introducing functional macromolecules into a variety of living cells is challenging but important for biology research and cell-based therapies. We report a novel cell delivery platform based on rotating shape anisotropic magnetic particles (SAMPs), which make very small cuts on cell membranes for macromolecule delivery with high efficiency and high survivability. SAMP delivery is performed by placing commercially available nickel powder onto cells grown in standard cell culture dishes. Application of a uniform magnetic field causes the magnetic particles to rotate because of mechanical torques induced by shape anisotropic magnetization. Cells touching these rotating particles are nicked, which generates transient membrane pores that enable the delivery of macromolecules into the cytosol of cells. Calcein dye, 3 and 40 kDa dextran polymers, a green fluorescence protein (GFP) plasmid, siRNA, and an enzyme (β-lactamase) were successfully delivered into HeLa cells, primary normal human dermal fibroblasts (NHDFs), and mouse cortical neurons that can be difficult to transfect. The SAMP approach offers several advantages, including easy implementation, low cost, high throughput, and efficient delivery of a broad range of macromolecules. Collectively, SAMP delivery has great potential for a broad range of academic and industrial applications.
Collapse
Affiliation(s)
- Ming-Yu Lin
- Instrument Technology Research Center, National Applied Research Laboratories, Hsinchu, Taiwan
| | - Yi-Chien Wu
- Mechanical and Aerospace Engineering Department, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ji-Ann Lee
- Department of Biological Chemistry, University of California at Los Angeles, Los Angeles, CA, USA
| | - Kuan-Wen Tung
- Mechanical and Aerospace Engineering Department, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jessica Zhou
- Mechanical and Aerospace Engineering Department, University of California at Los Angeles, Los Angeles, CA, USA
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA California NanoSystems Institute (CNSI), University of California at Los Angeles, Los Angeles, CA, USA
| | - J Andrew Yeh
- Instrument Technology Research Center, National Applied Research Laboratories, Hsinchu, Taiwan
| | - Pei Yu Chiou
- Mechanical and Aerospace Engineering Department, University of California at Los Angeles, Los Angeles, CA, USA Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, USA California NanoSystems Institute (CNSI), University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|