1
|
Nowak I, Madej M, Secemska J, Sarna R, Strzalka-Mrozik B. Virus-Based Biological Systems as Next-Generation Carriers for the Therapy of Central Nervous System Diseases. Pharmaceutics 2023; 15:1931. [PMID: 37514117 PMCID: PMC10384784 DOI: 10.3390/pharmaceutics15071931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/29/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Central nervous system (CNS) diseases are currently a major challenge in medicine. One reason is the presence of the blood-brain barrier, which is a significant limitation for currently used medicinal substances that are characterized by a high molecular weight and a short half-life. Despite the application of nanotechnology, there is still the problem of targeting and the occurrence of systemic toxicity. Viral vectors and virus-like particles (VLPs) may provide a promising solution to these challenges. Their small size, biocompatibility, ability to carry medicinal substances, and specific targeting of neural cells make them useful in research when formulating a new generation of biological carriers. Additionally, the possibility of genetic modification has the potential for gene therapy. Among the most promising viral vectors are adeno-associated viruses, adenoviruses, and retroviruses. This is due to their natural tropism to neural cells, as well as the possibility of genetic and surface modification. Moreover, VLPs that are devoid of infectious genetic material in favor of increasing capacity are also leading the way for research on new drug delivery systems. The aim of this study is to review the most recent reports on the use of viral vectors and VLPs in the treatment of selected CNS diseases.
Collapse
Affiliation(s)
- Ilona Nowak
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marcel Madej
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Julia Secemska
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Robert Sarna
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
2
|
Zhang Q, Liu J, Chen L, Zhang M. Promoting Endogenous Neurogenesis as a Treatment for Alzheimer's Disease. Mol Neurobiol 2023; 60:1353-1368. [PMID: 36445633 DOI: 10.1007/s12035-022-03145-2] [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: 06/01/2022] [Accepted: 11/19/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most universal neurodegenerative disorder characterized by memory loss and cognitive impairment. AD is biologically defined by production and aggregation of misfolded protein including extracellular amyloid β (Aβ) peptide and intracellular microtubule-associated protein tau tangles in neurons, leading to irreversible neuronal loss. At present, regulation of endogenous neurogenesis to supplement lost neurons has been proposed as a promising strategy for treatment of AD. However, the exact underlying mechanisms of impaired neurogenesis in AD have not been fully explained and effective treatments targeting neurogenesis for AD are limited. In this review, we mainly focus on the latest research of impaired neurogenesis in AD. Then we discuss the factors affecting stages of neurogenesis and the interplay between neural stem cells (NSCs) and neurogenic niche under AD pathological conditions. This review aims to explore potential therapeutic strategies that promote endogenous neurogenesis for AD treatments.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Jingyue Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China. .,School of Nursing, Jilin University, Changchun, China.
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
3
|
Wang X, Ma C, Rodríguez Labrada R, Qin Z, Xu T, He Z, Wei Y. Recent advances in lentiviral vectors for gene therapy. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1842-1857. [PMID: 34708326 DOI: 10.1007/s11427-021-1952-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Lentiviral vectors (LVs), derived from human immunodeficiency virus, are powerful tools for modifying the genes of eukaryotic cells such as hematopoietic stem cells and neural cells. With the extensive and in-depth studies on this gene therapy vehicle over the past two decades, LVs have been widely used in both research and clinical trials. For instance, third-generation and self-inactive LVs have been used to introduce a gene with therapeutic potential into the host genome and achieve targeted delivery into specific tissue. When LVs are employed in leukemia, the transduced T cells recognize and kill the tumor B cells; in β-thalassemia, the transduced CD34+ cells express normal β-globin; in adenosine deaminase-deficient severe combined immunodeficiency, the autologous CD34+ cells express adenosine deaminase and realize immune reconstitution. Overall, LVs can perform significant roles in the treatment of primary immunodeficiency diseases, hemoglobinopathies, B cell leukemia, and neurodegenerative diseases. In this review, we discuss the recent developments and therapeutic applications of LVs. The safe and efficient LVs show great promise as a tool for human gene therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuicui Ma
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Roberto Rodríguez Labrada
- Department Clinical Neurophysiology, Centre for the Research and Rehabilitation of Hereditary Ataxias, Holguín, 80100, Cuba
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Yuquan Wei
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Saleki K, Banazadeh M, Miri NS, Azadmehr A. Triangle of cytokine storm, central nervous system involvement, and viral infection in COVID-19: the role of sFasL and neuropilin-1. Rev Neurosci 2021; 33:147-160. [PMID: 34225390 DOI: 10.1515/revneuro-2021-0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) is identified as the cause of coronavirus disease 2019 (COVID-19), and is often linked to extreme inflammatory responses by over activation of neutrophil extracellular traps (NETs), cytokine storm, and sepsis. These are robust causes for multi-organ damage. In particular, potential routes of SARS-CoV2 entry, such as angiotensin-converting enzyme 2 (ACE2), have been linked to central nervous system (CNS) involvement. CNS has been recognized as one of the most susceptible compartments to cytokine storm, which can be affected by neuropilin-1 (NRP-1). ACE2 is widely-recognized as a SARS-CoV2 entry pathway; However, NRP-1 has been recently introduced as a novel path of viral entry. Apoptosis of cells invaded by this virus involves Fas receptor-Fas ligand (FasL) signaling; moreover, Fas receptor may function as a controller of inflammation. Furthermore, NRP-1 may influence FasL and modulate cytokine profile. The neuroimmunological insult by SARS-CoV2 infection may be inhibited by therapeutic approaches targeting soluble Fas ligand (sFasL), cytokine storm elements, or related viral entry pathways. In the current review, we explain pivotal players behind the activation of cytokine storm that are associated with vast CNS injury. We also hypothesize that sFasL may affect neuroinflammatory processes and trigger the cytokine storm in COVID-19.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, 76169-13555, Iran
| | - Niloufar Sadat Miri
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| | - Abbas Azadmehr
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- Medical Immunology Department, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| |
Collapse
|
5
|
Tedeschi DV, da Cunha AF, Cominetti MR, Pedroso RV. Efficacy of Gene Therapy to Restore Cognition in Alzheimer's Disease: A Systematic Review. Curr Gene Ther 2021; 21:246-257. [PMID: 33494678 DOI: 10.2174/1566523221666210120091146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/09/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the main cause of dementia and it is a progressive neurogenerative disease characterized by the accumulation of neurofibrillary tangles and senile plaques. There is currently no cure; however, some treatments are available to slow down the progression of the disease, including gene therapy, which has been investigated to have great potential for the treatment of AD. OBJECTIVE The aim of this review was to identify the efficacy of gene therapy to restore cognition in AD. METHODS A systematic review was carried out using papers published up to May 2020 and available in the Web of Science, Scopus, and Medline/PUBMED databases. Articles were considered for inclusion if they were original researches that investigated the effects of gene therapy on cognition in AD. The methodological quality of the selected studies was evaluated using the Risk of Bias Tool for Animal Intervention Studies (SYRCLE's Rob tool) and the Jadad Scale. RESULTS Most preclinical studies obtained positive results in improving memory and learning in mice that underwent treatment with gene therapy. On the other hand, clinical studies have obtained inconclusive results related to the delivery methods of the viral vector used in gene therapy. CONCLUSION Gene therapy has shown a great potential for the treatment of AD in preclinical trials, but results should be interpreted with caution since preclinical studies presented limitations to predict the efficacy of the treatment outcome in humans.
Collapse
Affiliation(s)
- Desyrre V Tedeschi
- Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| | - Anderson F da Cunha
- Department of Genetics and Evolution, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565- 905, Sao Carlos SP, Brazil
| | - Márcia R Cominetti
- Department of Gerontology, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| | - Renata Valle Pedroso
- Department of Gerontology, Federal University of Sao Carlos, Rodovia Washington Luis Km 310 - 13565-905, Sao Carlos SP, Brazil
| |
Collapse
|
6
|
Liu XH, Ning FB, Zhao DP, Chang YY, Wu HM, Zhang WH, Yu AL. Role of miR-211 in a PC12 cell model of Alzheimer's disease via regulation of neurogenin 2. Exp Physiol 2021; 106:1061-1071. [PMID: 33527539 DOI: 10.1113/ep088953] [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: 07/20/2020] [Accepted: 01/28/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the mechanism of miR-211 in an Alzheimer's disease cell model? What is the main finding and its importance? miR-211 was upregulated in an Alzheimer's disease cell model. It targeted neurogenin 2, reduced the activation of the phosphoinositide 3-kinase-Akt signalling pathway, inhibited the proliferation of the Alzheimer's disease cell model and promoted apoptosis. ABSTRACT MicroRNAs (miRs) are aberrantly expressed in Alzheimer's disease (AD) patients. This study was intended to investigate the effect of miR-211 on an AD cell model and the involvement of neurogenin 2 (Ngn2). The appropriate dose and time for the effect of Aβ1-42 on PC12 cells were determined to establish an AD cell model. An effect of miR-211 expression on cell viability, proliferation and apoptosis was detected after cell transfection. Online prediction and a dual luciferase reporter gene assay were utilized to confirm the binding sequence of miR-211 and Ngn2. qRT-PCR and western blot analysis were applied to measure Ngn2 expression. A gain and loss of function assay of miR-211 and Ngn2 was performed, and activation of the phosphoinositide 3-kinase (PI3K)-Akt signaling pathway was detected. The AD cell model was induced by Aβ1-42 treatment. miR-211 expression was significantly enhanced after miR-211 transfection, leading to suppressed proliferation and promotion of apoptosis in Aβ1-42 -treated PC12 cells. In addition, miR-211 could downregulate Ngn2 mRNA and protein expression, while overexpression of Ngn2 could reverse the effects of miR-211 on Aβ1-42 -treated PC12 cells and significantly enhance the phosphorylated Akt and PI3K protein levels. miR-211 could inhibit growth of PC12 cells by suppressing Ngn2 expression and inactivating the PI3K-Akt signalling pathway.
Collapse
Affiliation(s)
- Xin-Hong Liu
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Fang-Bo Ning
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Da-Peng Zhao
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Yan-Yan Chang
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Hua-Min Wu
- Department of Imaging, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Wen-Hiu Zhang
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Ai-Ling Yu
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| |
Collapse
|
7
|
Dou KX, Zhang C, Tan CC, Xu W, Li JQ, Cao XP, Tan L, Yu JT. Genome-wide association study identifies CBFA2T3 affecting the rate of CSF Aβ 42 decline in non-demented elders. Aging (Albany NY) 2019; 11:5433-5444. [PMID: 31370031 PMCID: PMC6710044 DOI: 10.18632/aging.102125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/21/2019] [Indexed: 11/30/2022]
Abstract
Brain amyloid deposition is an early pathological event in Alzheimer's disease (AD), and abnormally low levels amyloid-β42 peptide (Aβ42) in cerebrospinal fluid (CSF) can be detected in preclinical AD. To identify the genetic determinants that regulate the rate of CSF Aβ42 decline among non-demented elders, we conducted a genome-wide association study involved 321 non-demented elders from Alzheimer's Disease Neuroimaging Initiative (ADNI) 1/GO/2 cohorts restricted to non-Hispanic Caucasians. A novel genome-wide significant association of higher annualized percent decline of CSF Aβ42 in the gene CBFA2T3 (CBFA2/RUNX1 translocation partner 3; rs13333659-T; p = 2.24 × 10-9) was identified. Besides displaying abnormal CSF Aβ42 levels, rs13333659-T carriers were more likely to exhibit a greater longitudinal cognitive decline (p = 0.029, β = 0.097) and hippocampal atrophy (p = 0.029, β = -0.160) in the non-demented elders, especially for the participants who were amyloid-positive at baseline. These findings suggest rs13333659 in CBFA2T3 as a risk locus to modulate the decline rate of CSF Aβ42 preceding the onset of clinical symptoms.
Collapse
Affiliation(s)
- Kai-Xin Dou
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Can Zhang
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | |
Collapse
|
8
|
Loera-Valencia R, Piras A, Ismail MAM, Manchanda S, Eyjolfsdottir H, Saido TC, Johansson J, Eriksdotter M, Winblad B, Nilsson P. Targeting Alzheimer's disease with gene and cell therapies. J Intern Med 2018; 284:2-36. [PMID: 29582495 DOI: 10.1111/joim.12759] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) causes dementia in both young and old people affecting more than 40 million people worldwide. The two neuropathological hallmarks of the disease, amyloid beta (Aβ) plaques and neurofibrillary tangles consisting of protein tau are considered the major contributors to the disease. However, a more complete picture reveals significant neurodegeneration and decreased cell survival, neuroinflammation, changes in protein and energy homeostasis and alterations in lipid and cholesterol metabolism. In addition, gene and cell therapies for severe neurodegenerative disorders have recently improved technically in terms of safety and efficiency and have translated to the clinic showing encouraging results. Here, we review broadly current data within the field for potential targets that could modify AD through gene and cell therapy strategies. We envision that not only Aβ will be targeted in a disease-modifying treatment strategy but rather that a combination of treatments, possibly at different intervention times may prove beneficial in curing this devastating disease. These include decreased tau pathology, neuronal growth factors to support neurons and modulation of neuroinflammation for an appropriate immune response. Furthermore, cell based therapies may represent potential strategies in the future.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - A Piras
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M A M Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Neuro, Diseases of the Nervous System Patient Flow, Karolinska University Hospital, Huddinge, Sweden
| | - S Manchanda
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - H Eyjolfsdottir
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - T C Saido
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - J Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - P Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
9
|
Ulasov AV, Rosenkranz AA, Sobolev AS. Transcription factors: Time to deliver. J Control Release 2017; 269:24-35. [PMID: 29113792 DOI: 10.1016/j.jconrel.2017.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/17/2022]
Abstract
Transcription factors (TFs) are at the center of the broad regulatory network orchestrating gene expression programs that elicit different biological responses. For a long time, TFs have been considered as potent drug targets due to their implications in the pathogenesis of a variety of diseases. At the same time, TFs, located at convergence points of cellular regulatory pathways, are powerful tools providing opportunities both for cell type change and for managing the state of cells. This task formulation requires the TF modulation problem to come to the fore. We review several ways to manage TF activity (small molecules, transfection, nanocarriers, protein-based approaches), analyzing their limitations and the possibilities to overcome them. Delivery of TFs could revolutionize the biomedical field. Whether this forecast comes true will depend on the ability to develop convenient technologies for targeted delivery of TFs.
Collapse
Affiliation(s)
- Alexey V Ulasov
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Andrey A Rosenkranz
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia
| | - Alexander S Sobolev
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia.
| |
Collapse
|
10
|
Current Opinion on the Role of Neurogenesis in the Therapeutic Strategies for Alzheimer Disease, Parkinson Disease, and Ischemic Stroke; Considering Neuronal Voiding Function. Int Neurourol J 2016; 20:276-287. [PMID: 28043116 PMCID: PMC5209581 DOI: 10.5213/inj.1632776.388] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 12/12/2016] [Indexed: 01/01/2023] Open
Abstract
Neurological diseases such as Alzheimer, Parkinson, and ischemic stroke have increased in occurrence and become important health issues throughout the world. There is currently no effective therapeutic strategy for addressing neurological deficits after the development of these major neurological disorders. In recent years, it has become accepted that adult neural stem cells located in the subventricular and subgranular zones have the ability to proliferate and differentiate in order to replace lost or damaged neural cells. There have been many limitations in the clinical application of both endogenous and exogenous neurogenesis for neurological disorders. However, many studies have investigated novel mechanisms in neurogenesis and have shown that these limitations can potentially be overcome with appropriate stimulation and various approaches. We will review concepts related to possible therapeutic strategies focused on the perspective of neurogenesis for the treatment of patients diagnosed with Alzheimer disease, Parkinson disease, and ischemic stroke based on current reports.
Collapse
|
11
|
Chen Y, Pu J, Zhang B. Progress and Challenges of Cell Replacement Therapy for Neurodegenerative Diseases Based on Direct Neural Reprogramming. Hum Gene Ther 2016; 27:962-970. [PMID: 27589383 DOI: 10.1089/hum.2016.078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neurodegenerative diseases are characterized by protein aggregation and progressive degeneration of neurons, causing severe functional deficiency in cognition, behavior, and movement. Until now, there has been no effective treatment available in the clinic. Considering the selective loss of specific neurons in the human brain in the pathogenesis of these diseases, generating functional neurons in vitro or in vivo to replace the lost neurons represents a novel strategy to treat neurodegenerative diseases. Human embryonic stem cells and induced pluripotent stem cells have good potential for cell replacement therapy. However, limitations, such as the possibility of tumor formation, have hindered its applications. Recently, a novel approach, direct neural reprogramming, in which somatic cells are reprogrammed to functional neurons without a stem-cell state, has emerged an alternative for cell replacement. Specific human somatic cells can be reprogrammed to functional subtype neurons via the introduction of transcription factors, microRNAs, or small molecules in vitro and in vivo, thereby reducing the risk of carcinogenesis. Studies demonstrated symptomatic relief when induced neurons were transplanted into animal models. Although the direct neural reprogramming holds great promise for cell replacement therapy, there remain a number of challenges for its clinical application, including low efficiency, unclear mechanisms, and safety concerns. This review highlights the progress and challenges of this technique, and discusses perspectives for its applications in cell replacement.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology, 2nd Affiliated Hospital, School of Medicine, Zhejiang University , Zhejiang, People's Republic of China
| | - Jiali Pu
- Department of Neurology, 2nd Affiliated Hospital, School of Medicine, Zhejiang University , Zhejiang, People's Republic of China
| | - Baorong Zhang
- Department of Neurology, 2nd Affiliated Hospital, School of Medicine, Zhejiang University , Zhejiang, People's Republic of China
| |
Collapse
|