1
|
Zhao D, Hu M, Liu S. Glial cells in the mammalian olfactory bulb. Front Cell Neurosci 2024; 18:1426094. [PMID: 39081666 PMCID: PMC11286597 DOI: 10.3389/fncel.2024.1426094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The mammalian olfactory bulb (OB), an essential part of the olfactory system, plays a critical role in odor detection and neural processing. Historically, research has predominantly focused on the neuronal components of the OB, often overlooking the vital contributions of glial cells. Recent advancements, however, underscore the significant roles that glial cells play within this intricate neural structure. This review discus the diverse functions and dynamics of glial cells in the mammalian OB, mainly focused on astrocytes, microglia, oligodendrocytes, olfactory ensheathing cells, and radial glia cells. Each type of glial contributes uniquely to the OB's functionality, influencing everything from synaptic modulation and neuronal survival to immune defense and axonal guidance. The review features their roles in maintaining neural health, their involvement in neurodegenerative diseases, and their potential in therapeutic applications for neuroregeneration. By providing a comprehensive overview of glial cell types, their mechanisms, and interactions within the OB, this article aims to enhance our understanding of the olfactory system's complexity and the pivotal roles glial cells play in both health and disease.
Collapse
Affiliation(s)
| | | | - Shaolin Liu
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, Department of Biomedical Sciences, University of Georgia College of Veterinary Medicine, Athens, GA, United States
| |
Collapse
|
2
|
Marei HE, Khan MUA, Hasan A. Potential use of iPSCs for disease modeling, drug screening, and cell-based therapy for Alzheimer's disease. Cell Mol Biol Lett 2023; 28:98. [PMID: 38031028 PMCID: PMC10687886 DOI: 10.1186/s11658-023-00504-2] [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: 08/26/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic illness marked by increasing cognitive decline and nervous system deterioration. At this time, there is no known medication that will stop the course of Alzheimer's disease; instead, most symptoms are treated. Clinical trial failure rates for new drugs remain high, highlighting the urgent need for improved AD modeling for improving understanding of the underlying pathophysiology of disease and improving drug development. The development of induced pluripotent stem cells (iPSCs) has made it possible to model neurological diseases like AD, giving access to an infinite number of patient-derived cells capable of differentiating neuronal fates. This advance will accelerate Alzheimer's disease research and provide an opportunity to create more accurate patient-specific models of Alzheimer's disease to support pathophysiological research, drug development, and the potential application of stem cell-based therapeutics. This review article provides a complete summary of research done to date on the potential use of iPSCs from AD patients for disease modeling, drug discovery, and cell-based therapeutics. Current technological developments in AD research including 3D modeling, genome editing, gene therapy for AD, and research on familial (FAD) and sporadic (SAD) forms of the disease are discussed. Finally, we outline the issues that need to be elucidated and future directions for iPSC modeling in AD.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| | - Muhammad Umar Aslam Khan
- Biomedical Research Center, Qatar University, 2713, Doha, Qatar
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
3
|
Jeyaraman M, Rajendran RL, Muthu S, Jeyaraman N, Sharma S, Jha SK, Muthukanagaraj P, Hong CM, Furtado da Fonseca L, Santos Duarte Lana JF, Ahn BC, Gangadaran P. An update on stem cell and stem cell-derived extracellular vesicle-based therapy in the management of Alzheimer's disease. Heliyon 2023; 9:e17808. [PMID: 37449130 PMCID: PMC10336689 DOI: 10.1016/j.heliyon.2023.e17808] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/10/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Globally, neurological diseases pose a major burden to healthcare professionals in terms of the management and prevention of the disorder. Among neurological diseases, Alzheimer's disease (AD) accounts for 50%-70% of dementia and is the fifth leading cause of mortality worldwide. AD is a progressive, degenerative neurological disease, with the loss of neurons and synapses in the cerebral cortex and subcortical regions. The management of AD remains a debate among physicians as no standard and specific "disease-modifying" modality is available. The concept of 'Regenerative Medicine' is aimed at regenerating the degenerated neural tissues to reverse the pathology in AD. Genetically modified engineered stem cells modify the course of AD after transplantation into the brain. Extracellular vesicles (EVs) are an emerging new approach in cell communication that involves the transfer of cellular materials from parental cells to recipient cells, resulting in changes at the molecular and signaling levels in the recipient cells. EVs are a type of vesicle that can be transported between cells. Many have proposed that EVs produced from mesenchymal stem cells (MSCs) may have therapeutic promise in the treatment of AD. The biology of AD, as well as the potential applications of stem cells and their derived EVs-based therapy, were explored in this paper.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu, 600056, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Orthopedics, Government Dindigul Medical College and Hospital, Dindigul, Tamil Nadu, 624001, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Orthopedics, Shri Sathya Sai Medical College and Research Institute, Sri Balaji Vidyapeeth, Chengalpet, Tamil Nadu, 603108, India
| | - Shilpa Sharma
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Purushothaman Muthukanagaraj
- Department of Internal Medicine & Psychiatry, SUNY-Upstate Binghamton Clinical Campus, Binghamton, NY, 13904, USA
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Lucas Furtado da Fonseca
- Department of Orthopedics, The Federal University of São Paulo, São Paulo, 04023-062, SP, Brazil
| | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| |
Collapse
|
4
|
Martinez B, Peplow PV. Biomaterial and tissue-engineering strategies for the treatment of brain neurodegeneration. Neural Regen Res 2022; 17:2108-2116. [PMID: 35259816 PMCID: PMC9083174 DOI: 10.4103/1673-5374.336132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The incidence of neurodegenerative diseases is increasing due to changing age demographics and the incidence of sports-related traumatic brain injury is tending to increase over time. Currently approved medicines for neurodegenerative diseases only temporarily reduce the symptoms but cannot cure or delay disease progression. Cell transplantation strategies offer an alternative approach to facilitating central nervous system repair, but efficacy is limited by low in vivo survival rates of cells that are injected in suspension. Transplanting cells that are attached to or encapsulated within a suitable biomaterial construct has the advantage of enhancing cell survival in vivo. A variety of biomaterials have been used to make constructs in different types that included nanoparticles, nanotubes, microspheres, microscale fibrous scaffolds, as well as scaffolds made of gels and in the form of micro-columns. Among these, Tween 80-methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles loaded with rhynchophylline had higher transport across a blood-brain barrier model and decreased cell death in an in vitro model of Alzheimer’s disease than rhynchophylline or untreated nanoparticles with rhynchophylline. In an in vitro model of Parkinson’s disease, trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin had a similar protective ability as free non-Fe hemin. A positive effect on neuron survival in several in vivo models of Parkinson’s disease was associated with the use of biomaterial constructs such as trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin, carbon nanotubes with olfactory bulb stem cells, poly(lactic-co-glycolic acid) microspheres with attached DI-MIAMI cells, ventral midbrain neurons mixed with short fibers of poly-(L-lactic acid) scaffolds and reacted with xyloglucan with/without glial-derived neurotrophic factor, ventral midbrain neurons mixed with Fmoc-DIKVAV hydrogel with/without glial-derived neurotrophic factor. Further studies with in vivo models of Alzheimer’s disease and Parkinson’s disease are warranted especially using transplantation of cells in agarose micro-columns with an inner lumen filled with an appropriate extracellular matrix material.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
5
|
Rezk S, Lashen S, El-Adl M, Elshopakey GE, Elghareeb MM, Hendam BM, Caceci T, Cenciarelli C, Marei HE. Effects of Rosemary Oil (Rosmarinus officinalis) supplementation on the fate of the transplanted human olfactory bulb neural stem cells against ibotenic acid-induced neurotoxicity (Alzheimer model) in rat. Metab Brain Dis 2022; 37:973-988. [PMID: 35075502 DOI: 10.1007/s11011-021-00890-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/05/2021] [Indexed: 12/16/2022]
Abstract
Rosemary oil (ROO) is known to have multiple pharmacological effects: it is an antioxidant, anti-inflammatory, and cytoprotective. In the present study, we examined the effects of ROO on Human olfactory bulb neuronal stem cells (hOBNSCs) after their transplantation into rats, with the ibotenic (IBO) acid-induced cognitive deficit model. After 7 weeks, cognitive functions were assessed using the Morris water maze (MWM). After two months blood and hippocampus samples were collected for biochemical, gene expression, and histomorphometric analyses. Learning ability and memory function were significantly enhanced (P < 0.05) after hOBNSCs transplantation and were nearly returned to normal in the treated group. The IBO acid injection was associated with a significant decline (P < 0.05) of total leukocyte count (TLC) and a significant increase (P < 0.05) in total and toxic neutrophils. As well, the level of IL-1β, TNF-α CRP in serum and levels of MDA and NO in hippocampus tissue were significantly elevated (P < 0.05), while antioxidant markers (CAT, GSH, and SOD) were reduced (P < 0.05) in treated tissue compared to controls. The administration of ROO before or with cell transplantation attenuated all these parameters. In particular, the level of NO nearly returned to normal when rosemary was administrated before cell transplantation. Gene expression analysis revealed the potential protective effect of ROO and hOBNSCs via down-expression of R-βAmyl and R- CAS 3 and R-GFAP genes. The improvement in the histological organization of the hippocampus was detected after the hOBNSCs transplantation especially in h/ROO/hOBNSCs group.
Collapse
Affiliation(s)
- Shaymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Samah Lashen
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Adl
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Gehad E Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | - Mona M Elghareeb
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Basma M Hendam
- Department of Husbandry & Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Thomas Caceci
- Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine , Blacksburg, VA, USA
| | - Carlo Cenciarelli
- Departament of Biomedical Sciences, Institute of Translational Pharmacology-CNR, Rome, Italy
| | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
6
|
Qin C, Li Y, Wang K. Functional Mechanism of Bone Marrow-Derived Mesenchymal Stem Cells in the Treatment of Animal Models with Alzheimer's Disease: Inhibition of Neuroinflammation. J Inflamm Res 2021; 14:4761-4775. [PMID: 34566422 PMCID: PMC8456430 DOI: 10.2147/jir.s327538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/18/2021] [Indexed: 12/27/2022] Open
Abstract
The transplantation of bone marrow-derived mesenchymal stem cells (BMMSCs) alleviates neuropathology and improves cognitive deficits in animal models with Alzheimer’s disease. However, the underlying mechanisms remain to be determined. Available data demonstrate transplanted BMMSCs can inhibit neuroinflammation, which may be related to microglial M1/M2 polarization and is regulated by the secretion of autocrine and paracrine cytokines. BMMSCs also mitigate Aβ plaques and Tau tangles in the brain, which may be associated with the recruitment of peripheral blood monocytes and the subsequent comprehensive effects. The therapeutic effects of stem cells involve potential mechanisms such as immunomodulation, apoptosis, and proliferation. BMMSC-mediated functional reconstruction through dynamic remodeling develops a novel balance. Herein, present review recapitulates the molecular basis of BMMSC-assisted biological processes and summarizes the possible mechanisms related to the interaction between BMMSCs and microglia. The transplanted BMMSCs can suppress neuroinflammation that plays a key role in the pathogenesis of Alzheimer’s disease.
Collapse
Affiliation(s)
- Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yongning Li
- Department of International Medical Service & Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Kewei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
7
|
Lin MS, Chiu IH, Lin CC. Ultrarapid Inflammation of the Olfactory Bulb After Spinal Cord Injury: Protective Effects of the Granulocyte Colony-Stimulating Factor on Early Neurodegeneration in the Brain. Front Aging Neurosci 2021; 13:701702. [PMID: 34248610 PMCID: PMC8267925 DOI: 10.3389/fnagi.2021.701702] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
The correlation among olfactory dysfunction, spinal cord injury (SCI), subjective cognitive decline, and neurodegenerative dementia has been established. Impaired olfaction is considered a marker for neurodegeneration. Hence, there is a need to examine if SCI leads to olfactory dysfunction. In this study, the brain tissue of mice with spinal cord hemisection injury was subjected to microarray analysis. The mRNA expression levels of olfactory receptors in the brain began to decline at 8 h post-SCI. SCI promoted neuroinflammation, downregulated the expression of olfactory receptors, decreased the number of neural stem cells (NSCs), and inhibited the production of neurotrophic factors in the olfactory bulbs at 8 h post-SCI. In particular, the SCI group had upregulated mRNA and protein expression levels of glial fibrillary acidic protein (GFAP; a marker of astrocyte reactivation) and pro-inflammatory mediators [IL-1β, IL-6, and Nestin (marker of NSCs)] in the olfactory bulb compared to levels in the sham control group. The mRNA expression levels of olfactory receptors (Olfr1494, Olfr1324, Olfr1241, and Olfr979) and neurotrophic factors [brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), and nerve growth factor (NGF)] were downregulated in the olfactory bulb of the SCI group mice at 8 h post-SCI. The administration of granulocyte colony-stimulating factor (G-CSF) mitigated these SCI-induced pathological changes in the olfactory bulb at 8 h post-SCI. These results indicate that the olfactory bulb is vulnerable to environmental damage even if the lesion is located at sites distant from the brain, such as the spinal cord. Additionally, SCI initiated pathological processes, including inflammatory response, and impaired neurogenesis, at an early stage. The findings of this study will provide a basis for future studies on pathological mechanisms of early neurodegenerative diseases involving the olfactory bulb and enable early clinical drug intervention.
Collapse
Affiliation(s)
- Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung, Taiwan.,Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan, Taiwan.,Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung, Taiwan.,Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung, Taiwan
| | - I-Hsiang Chiu
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan, Taiwan
| | - Chai-Ching Lin
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan, Taiwan
| |
Collapse
|
8
|
Defteralı Ç, Moreno-Estellés M, Crespo C, Díaz-Guerra E, Díaz-Moreno M, Vergaño-Vera E, Nieto-Estévez V, Hurtado-Chong A, Consiglio A, Mira H, Vicario C. Neural stem cells in the adult olfactory bulb core generate mature neurons in vivo. Stem Cells 2021; 39:1253-1269. [PMID: 33963799 DOI: 10.1002/stem.3393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 04/20/2021] [Indexed: 01/05/2023]
Abstract
Although previous studies suggest that neural stem cells (NSCs) exist in the adult olfactory bulb (OB), their location, identity, and capacity to generate mature neurons in vivo has been little explored. Here, we injected enhanced green fluorescent protein (EGFP)-expressing retroviral particles into the OB core of adult mice to label dividing cells and to track the differentiation/maturation of any neurons they might generate. EGFP-labeled cells initially expressed adult NSC markers on days 1 to 3 postinjection (dpi), including Nestin, GLAST, Sox2, Prominin-1, and GFAP. EGFP+ -doublecortin (DCX) cells with a migratory morphology were also detected and their abundance increased over a 7-day period. Furthermore, EGFP-labeled cells progressively became NeuN+ neurons, they acquired neuronal morphologies, and they became immunoreactive for OB neuron subtype markers, the most abundant representing calretinin expressing interneurons. OB-NSCs also generated glial cells, suggesting they could be multipotent in vivo. Significantly, the newly generated neurons established and received synaptic contacts, and they expressed presynaptic proteins and the transcription factor pCREB. By contrast, when the retroviral particles were injected into the subventricular zone (SVZ), nearly all (98%) EGFP+ -cells were postmitotic when they reached the OB core, implying that the vast majority of proliferating cells present in the OB are not derived from the SVZ. Furthermore, we detected slowly dividing label-retaining cells in this region that could correspond to the population of resident NSCs. This is the first time NSCs located in the adult OB core have been shown to generate neurons that incorporate into OB circuits in vivo.
Collapse
Affiliation(s)
- Çağla Defteralı
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mireia Moreno-Estellés
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain.,Instituto de Biomedicina de Valencia-CSIC (IBV-CSIC), Valencia, Spain
| | - Carlos Crespo
- Departamento de Biología Celular, Estructura de Investigación Interdisciplinar en Biotecnología y Biomedicina (BIOTECMED), Universitat de Valencia, Valencia, Spain
| | - Eva Díaz-Guerra
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Díaz-Moreno
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain
| | - Eva Vergaño-Vera
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Vanesa Nieto-Estévez
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anahí Hurtado-Chong
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Antonella Consiglio
- Institute of Biomedicine, Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Helena Mira
- Unidad de Neurobiología Molecular, Área de Biología Celular y del Desarrollo, CNM-ISCIII, Majadahonda, Spain.,Instituto de Biomedicina de Valencia-CSIC (IBV-CSIC), Valencia, Spain
| | - Carlos Vicario
- Instituto Cajal-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,CIBERNED-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
9
|
Ahani-Nahayati M, Shariati A, Mahmoodi M, Olegovna Zekiy A, Javidi K, Shamlou S, Mousakhani A, Zamani M, Hassanzadeh A. Stem cell in neurodegenerative disorders; an emerging strategy. Int J Dev Neurosci 2021; 81:291-311. [PMID: 33650716 DOI: 10.1002/jdn.10101] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 01/28/2023] Open
Abstract
Neurodegenerative disorders are a diversity of disorders, surrounding Alzheimer's (AD), Parkinson's (PD), Huntington's diseases (HD), and amyotrophic lateral sclerosis (ALS) accompanied by some other less common diseases generally characterized by either developed deterioration of central or peripheral nervous system structurally or functionally. Today, with the viewpoint of an increasingly aging society, the number of patients with neurodegenerative diseases and sociomedical burdens will spread intensely. During the last decade, stem cell technology has attracted great attention for treating neurodegenerative diseases worldwide because of its unique attributes. As acknowledged, there are several categories of stem cells being able to proliferate and differentiate into various cellular lineages, highlighting their significance in the context of regenerative medicine. In preclinical models, stem cell therapy using mesenchymal stem/stromal cells (MSCs), hematopoietic stem cells (HSCs), and neural progenitor or stem cells (NPCs or NSCs) along with pluripotent stem cells (PSCs)-derived neuronal cells could elicit desired therapeutic effects, enabling functional deficit rescue partially. Regardless of the noteworthy progress in our scientific awareness and understanding of stem cell biology, there still exist various challenges to defeat. In the present review, we provide a summary of the therapeutic potential of stem cells and discuss the current status and prospect of stem cell strategy in neurodegenerative diseases, in particular, AD, PD, ALS, and HD.
Collapse
Affiliation(s)
- Milad Ahani-Nahayati
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ali Shariati
- Stem Cell Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahnaz Mahmoodi
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kamran Javidi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Shamlou
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Akbar Mousakhani
- Department of Plant Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Neurosciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Kahroba H, Ramezani B, Maadi H, Sadeghi MR, Jaberie H, Ramezani F. The role of Nrf2 in neural stem/progenitors cells: From maintaining stemness and self-renewal to promoting differentiation capability and facilitating therapeutic application in neurodegenerative disease. Ageing Res Rev 2021; 65:101211. [PMID: 33186670 DOI: 10.1016/j.arr.2020.101211] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDs) cause progressive loss of neurons in nervous system. NDs are categorized as acute NDs such as stroke and head injury, besides chronic NDs including Alzheimer's, Parkinson's, Huntington's diseases, Friedreich's Ataxia, Multiple Sclerosis. The exact etiology of NDs is not understood but oxidative stress, inflammation and synaptic dysfunction are main hallmarks. Oxidative stress leads to free radical attack on neural cells which contributes to protein misfolding, glia cell activation, mitochondrial dysfunction, impairment of DNA repair system and subsequently cellular death. Neural stem cells (NSCs) support adult neurogenesis in nervous system during injuries which is limited to certain regions in brain. NSCs can differentiate into the neurons, astrocytes or oligodendrocytes. Impaired neurogenesis and inadequate induction of neurogenesis are the main obstacles in treatment of NDs. Protection of neural cells from oxidative damages and supporting neurogenesis are promising strategies to treat NDs. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a transcriptional master regulator that maintains the redox homeostasis in cells by provoking expression of antioxidant, anti-inflammatory and cytoprotective genes. Nrf2 can strongly influence the NSCs function and fate determination by reducing levels of reactive oxygen species in benefit of NSC survival and neurogenesis. In this review we will summarize the role of Nrf2 in NSC function, and exogenous and endogenous therapeutic strategies in treatment of NDs.
Collapse
Affiliation(s)
- Houman Kahroba
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Ramezani
- Department of Chemistry, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Hamid Maadi
- Department of Medical Genetics, and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Jaberie
- Department of Biochemistry, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Fatemeh Ramezani
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Non-viral delivery systems of DNA into stem cells: Promising and multifarious actions for regenerative medicine. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
The Fate of Transplanted Olfactory Progenitors Is Conditioned by the Cell Phenotypes of the Receiver Brain Tissue in Cocultures. Int J Mol Sci 2020; 21:ijms21197249. [PMID: 33008128 PMCID: PMC7582579 DOI: 10.3390/ijms21197249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Among the numerous candidates for cell therapy of the central nervous system (CNS), olfactory progenitors (OPs) represent an interesting alternative because they are free of ethical concerns, are easy to collect, and allow autologous transplantation. In the present study, we focused on the optimization of neuron production and maturation. It is known that plated OPs respond to various trophic factors, and we also showed that the use of Nerve Growth Factor (NGF) allowed switching from a 60/40 neuron/glia ratio to an 80/20 one. Nevertheless, in order to focus on the integration of OPs in mature neural circuits, we cocultured OPs in primary cultures obtained from the cortex and hippocampus of newborn mice. When dissociated OPs were plated, they differentiated into both glial and neuronal phenotypes, but we obtained a 1.5-fold higher viability in cortex/OP cocultures than in hippocampus/OP ones. The fate of OPs in cocultures was characterized with different markers such as BrdU, Map-2, and Synapsin, indicating a healthy integration. These results suggest that the integration of transplanted OPs might by affected by trophic factors and the environmental conditions/cell phenotypes of the host tissue. Thus, a model of coculture could provide useful information on key cell events for the use of progenitors in cell therapy.
Collapse
|
13
|
Madl CM, LeSavage BL, Khariton M, Heilshorn SC. Neural Progenitor Cells Alter Chromatin Organization and Neurotrophin Expression in Response to 3D Matrix Degradability. Adv Healthc Mater 2020; 9:e2000754. [PMID: 32743903 DOI: 10.1002/adhm.202000754] [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: 05/04/2020] [Revised: 07/14/2020] [Indexed: 11/09/2022]
Abstract
Neural progenitor cells (NPCs) are promising therapeutic candidates for nervous system regeneration. Significant efforts focus on developing hydrogel-based approaches to facilitate the clinical translation of NPCs, from scalable platforms for stem cell production to injectable carriers for cell transplantation. However, fundamental questions surrounding NPC-hydrogel interactions remain unanswered. While matrix degradability is known to regulate the stemness and differentiation capacity of NPCs, how degradability impacts NPC epigenetic regulation and secretory phenotype remains unknown. To address this question, NPCs encapsulated in recombinant protein hydrogels with tunable degradability are assayed for changes in chromatin organization and neurotrophin expression. In high degradability gels, NPCs maintain expression of stem cell factors, proliferate, and have large nuclei with elevated levels of the stemness-associated activating histone mark H3K4me3. In contrast, NPCs in low degradability gels exhibit more compact, rounded nuclei with peripherally localized heterochromatin, are non-proliferative yet non-senescent, and maintain expression of neurotrophic factors with potential therapeutic relevance. This work suggests that tuning matrix degradability may be useful to direct NPCs toward either a more-proliferative, stem-like phenotype for cell replacement therapies, or a more quiescent-like, pro-secretory phenotype for soluble factor-mediated therapies.
Collapse
Affiliation(s)
- Christopher M. Madl
- Department of Bioengineering Stanford University Stanford CA 94305 USA
- Baxter Laboratory for Stem Cell Biology Department of Microbiology & Immunology Stanford University Stanford CA 94305 USA
| | - Bauer L. LeSavage
- Department of Bioengineering Stanford University Stanford CA 94305 USA
| | | | - Sarah C. Heilshorn
- Department of Materials Science & Engineering Stanford University 476 Lomita Mall, McCullough Room 246 Stanford CA 94305 USA
| |
Collapse
|
14
|
Qin C, Lu Y, Wang K, Bai L, Shi G, Huang Y, Li Y. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer's disease: a meta-analytic review on potential mechanisms. Transl Neurodegener 2020; 9:20. [PMID: 32460886 PMCID: PMC7251864 DOI: 10.1186/s40035-020-00199-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative disorder. Therapeutically, a transplantation of bone marrow mesenchymal stem cells (BMMSCs) can play a beneficial role in animal models of Alzheimer's disease. However, the relevant mechanism remains to be fully elucidated. MAIN BODY Subsequent to the transplantation of BMMSCs, memory loss and cognitive impairment were significantly improved in animal models with Alzheimer's disease (AD). Potential mechanisms involved neurogenesis, apoptosis, angiogenesis, inflammation, immunomodulation, etc. The above mechanisms might play different roles at certain stages. It was revealed that the transplantation of BMMSCs could alter some gene levels. Moreover, the differential expression of representative genes was responsible for neuropathological phenotypes in Alzheimer's disease, which could be used to construct gene-specific patterns. CONCLUSIONS Multiple signal pathways involve therapeutic mechanisms by which the transplantation of BMMSCs improves cognitive and behavioral deficits in AD models. Gene expression profile can be utilized to establish statistical regression model for the evaluation of therapeutic effect. The transplantation of autologous BMMSCs maybe a prospective therapy for patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China.
| | - Yalan Lu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Kewei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Lin Bai
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Guiying Shi
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yiying Huang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yongning Li
- Department of International Medical Service & Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan 1, Dong Cheng District, Beijing, 100730, China
| |
Collapse
|
15
|
Poltavtseva RA, Samokhin AN, Bobkova NV, Alexandrova MA, Sukhikh GT. Effect of Transplantation of Neural Stem and Progenitor Cells on Memory in Animals with Alzheimer's Type Neurodegeneration. Bull Exp Biol Med 2020; 168:589-596. [PMID: 32152851 DOI: 10.1007/s10517-020-04758-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Indexed: 12/14/2022]
Abstract
The effects of systemic and intracerebral transplantation of human fetal neural stem and progenitor cells were studied on the model of olfactory bulbectomy in mice with developing signs of sporadic Alzheimer's disease. It was found that transplantation of these cells at certain stages of disease development contributed to improvement of spatial memory and preservation of hippocampal neurons in these animals.
Collapse
Affiliation(s)
- R A Poltavtseva
- V. I. Kulakov National Medical Research Center for Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A N Samokhin
- Institute of Cell Biophysics, Federal research Center Pushchino Research Center of Biological Studies, Russian Academy of Science, Pushchino, Moscow region, Russia
| | - N V Bobkova
- Institute of Cell Biophysics, Federal research Center Pushchino Research Center of Biological Studies, Russian Academy of Science, Pushchino, Moscow region, Russia
| | - M A Alexandrova
- N. K. Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - G T Sukhikh
- V. I. Kulakov National Medical Research Center for Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
16
|
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease caused by eventually aggregated amyloid β (Aβ) plaques in degenerating neurons of the aging brain. These aggregated protein plaques mainly consist of Aβ fibrils and neurofibrillary tangles (NFTs) of phosphorylated tau protein. Even though some cholinesterase inhibitors, NMDA receptor antagonist, and monoclonal antibodies were developed to inhibit neurodegeneration or activate neural regeneration or clear off the Aβ deposits, none of the treatment is effective in improving the cognitive and memory dysfunctions of the AD patients. Thus, stem cell therapy represents a powerful tool for the treatment of AD. In addition to discussing the advents in molecular pathogenesis and animal models of this disease and the treatment approaches using small molecules and immunoglobulins against AD, we will focus on the stem cell sources for AD using neural stem cells (NSCs); embryonic stem cells (ESCs); and mesenchymal stem cells (MSCs) from bone marrow, umbilical cord, and umbilical cord blood. In particular, patient-specific-induced pluripotent stem cells (iPS cells) are proposed as a future prospective and the challenges for the treatment of AD.
Collapse
|
17
|
Han F, Liu C, Huang J, Chen J, Wei C, Geng X, Liu Y, Han D, Li M. The application of patient-derived induced pluripotent stem cells for modeling and treatment of Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2019. [DOI: 10.1177/2096595819896178] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent age-related neurodegenerative disease which is mainly caused by aggregated protein plaques in degenerating neurons of the brain. These aggregated protein plaques are mainly consisting of amyloid β (Aβ) fibrils and neurofibrillary tangles (NFTs) of phosphorylated tau protein. Even though the transgenic murine models can recapitulate some of the AD phenotypes, they are not the human cell models of AD. Recent breakthrough in somatic cell reprogramming made it available to use induced pluripotent stem cells (iPSCs) for patientspecific disease modeling and autologous transplantation therapy. Human iPSCs provide alternative ways to obtain specific human brain cells of AD patients to study the molecular mechanisms and therapeutic approaches for familial and sporadic forms of AD. After differentiation into neuronal cells, iPSCs have enabled the investigation of the complex aetiology and timescale over which AD develops in human brain. Here, we first go over the pathological process of and transgenic models of AD. Then we discuss the application of iPSC for disease model and cell transplantation. At last the challenges and future applications of iPSCs for AD will be summarized to propose cell-based approaches for the treatment of this devastating disorder.
Collapse
Affiliation(s)
- Fabin Han
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Shandong 252000, China
- The Translational Research Lab for Stem Cells and Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
- These authors contributed equally to this work
| | - Chuanguo Liu
- The Translational Research Lab for Stem Cells and Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
- These authors contributed equally to this work
| | - Jin Huang
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, The Fourth Medical Centre of PLA General Hospital, Beijing 100048, China
- These authors contributed equally to this work
| | - Juanli Chen
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Shandong 252000, China
| | - Chuanfei Wei
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Shandong 252000, China
| | - Xiwen Geng
- The Translational Research Lab for Stem Cells and Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Yanming Liu
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Shandong 252000, China
| | - Dong Han
- Shandong Molecular Diagnostics & Cell Therapeutic Biotechnology Corporation, Ji’nan, Shandong 250001, China
| | - Mengpeng Li
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Shandong 252000, China
| |
Collapse
|
18
|
Han F, Liu C, Huang J, Chen J, Wei C, Geng X, Liu Y, Han D, Li M. The application of patient-derived induced pluripotent stem cells for modeling and treatment of Alzheimer’s disease. BRAIN SCIENCE ADVANCES 2019. [DOI: 10.26599/bsa.2019.9050003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
19
|
Crane AT, Voth JP, Shen FX, Low WC. Concise Review: Human-Animal Neurological Chimeras: Humanized Animals or Human Cells in an Animal? Stem Cells 2019; 37:444-452. [PMID: 30629789 DOI: 10.1002/stem.2971] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 12/03/2018] [Indexed: 12/24/2022]
Abstract
Blastocyst complementation is an emerging methodology in which human stem cells are transferred into genetically engineered preimplantation animal embryos eventually giving rise to fully developed human tissues and organs within the animal host for use in regenerative medicine. The ethical issues surrounding this method have caused the National Institutes of Health to issue a moratorium on funding for blastocyst complementation citing the potential for human cells to substantially contribute to the brain of the chimeric animal. To address this concern, we performed an in-depth review of the neural transplantation literature to determine how the integration of human cells into the nonhuman neural circuitry has altered the behavior of the host. Despite reports of widespread integration of human cell transplants, our review of 150 transplantation studies found no evidence suggestive of humanization of the animal host, and we thus conclude that, at present, concerns over humanization should not prevent research on blastocyst complementation to continue. We suggest proceeding in a controlled and transparent manner, however, and include recommendations for future research with careful consideration for how human cells may contribute to the animal host nervous system. Stem Cells 2019;37:444-452.
Collapse
Affiliation(s)
- Andrew T Crane
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Minnesota Craniofacial Research Training Program, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph P Voth
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Francis X Shen
- University of Minnesota Law School, Minneapolis, Minnesota, USA.,Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Walter C Low
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA.,Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
20
|
Marei HE, Casalbore P, Althani A, Coccè V, Cenciarelli C, Alessandri G, Brini AT, Parati E, Bondiolotti G, Pessina A. Human Olfactory Bulb Neural Stem Cells (Hu-OBNSCs) Can Be Loaded with Paclitaxel and Used to Inhibit Glioblastoma Cell Growth. Pharmaceutics 2019; 11:pharmaceutics11010045. [PMID: 30669623 PMCID: PMC6358986 DOI: 10.3390/pharmaceutics11010045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Exploitation of the potential ability of human olfactory bulb (hOB) cells to carry, release, and deliver an effective, targeted anticancer therapy within the central nervous system (CNS) milieu remains elusive. Previous studies have demonstrated the marked ability of several types of stem cells (such as mesenchymal stem cells (MSCs) to carry and release different anti-cancer agents such as paclitaxel (PTX). Herein we investigate the ability of human olfactory bulb neural stem cells (Hu-OBNSCs) to carry and release paclitaxel, producing effective cytotoxic effects against cancer cells. We isolated Hu-OBNSCs from the hOB, uploaded them with PTX, and studied their potential cytotoxic effects against cancer cells in vitro. Interestingly, the Hu-OBNSCs displayed a five-fold increase in their resistance to the cytotoxicity of PTX, and the PTX-uploaded Hu-OBNSCs were able to inhibit proliferation and invasion, and to trigger marked cytotoxic effects on glioblastoma multiforme (GBM) cancer cells, and Human Caucasian fetal pancreatic adenocarcinoma 1 (CFPAC-1) in vitro. Despite their ability to resist the cytotoxic activity of PTX, the mechanism by which Hu-OBNSCs acquire resistance to PTX is not yet explained. Collectively our data indicate the ability of the Hu-OBNSCs to resist PTX, and to trigger effective cytotoxic effects against GBM cancer cells and CFPAC-1. This indicates their potential to be used as a carrier/vehicle for targeted anti-cancer therapy within the CNS.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Patrizia Casalbore
- Institute of Cell Biology and Neurobiology, National Research Council of Italy, 00015 Rome, Italy.
| | - Asmaa Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Valentina Coccè
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council of Italy, 00133 Rome, Italy.
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, 20133 Milan, Italy.
| | - Anna T Brini
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy.
| | - Eugenio Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, 20133 Milan, Italy.
| | - Gianpietro Bondiolotti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy.
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
21
|
Alipour M, Nabavi SM, Arab L, Vosough M, Pakdaman H, Ehsani E, Shahpasand K. Stem cell therapy in Alzheimer's disease: possible benefits and limiting drawbacks. Mol Biol Rep 2018; 46:1425-1446. [PMID: 30565076 DOI: 10.1007/s11033-018-4499-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death globally and the main reason for dementia in elderly people. AD is a long-term and progressive neurodegenerative disorder that steadily worsens memory and communicating skills eventually leads to a disabled person of performing simple daily tasks. Unfortunately, numerous clinical trials exploring new therapeutic drugs have encountered disappointing outcomes in terms of improved cognitive performance since they are not capable of halting or stimulating the regeneration of already-damaged neural cells, and merely provide symptomatic relief. Therefore, a deeper understanding of the mechanism of action of stem cell may contribute to the development of novel and effective therapies. The revolutionary discovery of stem cells has cast a new hope for the development of disease-modifying treatments for AD, in terms of their potency in the replenishment of lost cells via differentiating towards specific lineages, stimulating in situ neurogenesis, and delivering the therapeutic agents to the brain. Herein, firstly, we explore the pathophysiology of AD. Next, we summarize the most recent preclinical stem cell reports designed for AD treatment, their benefits and outcomes according to cell type. We briefly review relevant clinical trials and their potential clinical applications in order to find a unique solution to effectively relieve the patients' pain.
Collapse
Affiliation(s)
- Masoume Alipour
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Leila Arab
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Ehsani
- Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Banihashem Sq., Banihashem St., Resalat highway, P.O. Box 19395-4644, Tehran, Iran.
| |
Collapse
|
22
|
Zhang F, Chen SQ, Tong MM, Wang PJ, Teng GJ. 7.0 tesla high resolution MRI study on intracerebral migration of magnet-labeled neural stem cells in a mouse model of Alzheimer's disease. Magn Reson Imaging 2018; 54:58-62. [PMID: 30118826 DOI: 10.1016/j.mri.2018.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To observe the migration characteristics of neural stem cells (NSCs) labeled with the MRI contrast agent superparamagnetic iron oxide (SPIO) in the brain of APP/PS1 transgenic mice with Alzheimer's disease (AD) by 7.0 T high resolution MRI. METHODS C57BL/6 mouse NSCs were cultured, amplified, labeled with Feridex and Poly-l-lysine (FE-PLL) and evaluated by transmission electron microscopy (TEM). Using the random number table method, 24 APP/PS1 transgenic AD mice aged 12 months were equally assigned to two groups: animals in group A were transplanted with FE-PLL labeled NSCs and those in group B were transplanted with non-labeled NSCs in the right hippocampus. Twelve wild-type mice of the same age and born from the same litter were used as the control group (group C) and transplanted with FE-PLL labeled NSCs. Using the 7.0 T high resolution MR scanner, the transplanted NSCs were traced in vivo at 1 day, 1 and 2 weeks after cell transplantation. The MRI findings were compared with the histopathological findings. RESULTS C57BL/6 mouse NSCs were cultured and amplified successfully. TEM showed large amounts of iron-containing particles in the cytoplasm of transplanted cells. MRI in group A showed the presence of spheroid low signals at the injection point of the hippocampus on T2*WI one day after transplantation; one weeks later, the low signals were seen diffusing to the surroundings along the injection point, and covering almost the whole hippocampal area but the intensity of the low signals became weaker gradually; two weeks after transplantation, almost all low signals disappeared. In group B, no significant change in low signals was observed in the transplantation area at all designated time points. Although low signals were also observed in the hippocampus after transplantation of FE-PLL labeled NSCs in group C, their size and location remained almost unchanged. Prussian blue staining showed that migration of the FE-PLL labeled NSCs in the hippocampus of the AD mice was consistent with the MRI findings at all designated time points. CONCLUSION NSCs underwent diffuse and non-directional migration to the surroundings after they were transplanted to the hippocampus of APP/PS1 transgenic AD mice, and this migration pattern could be traced in vivo by MRI when they were labeled with magnet.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China
| | - Shuang-Qing Chen
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China.
| | - Ming-Min Tong
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China
| | - Pei-Jun Wang
- Department of Radiology, the Affiliated Tongji Hospital of Tongji University, Shanghai 200065, China
| | - Gao-Jun Teng
- Key Laboratory of Molecular Imaging, Southeast University, Nanjing 210009, China
| |
Collapse
|
23
|
Hoveizi E, Mohammadi T, Moazedi AA, Zamani N, Eskandary A. Transplanted neural-like cells improve memory and Alzheimer-like pathology in a rat model. Cytotherapy 2018; 20:964-973. [PMID: 30025963 DOI: 10.1016/j.jcyt.2018.03.036] [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: 01/24/2018] [Revised: 03/18/2018] [Accepted: 03/30/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND AIMS Degeneration of the central nerve system, particularly in Alzheimer's disease, is a burden on society, and despite years of research, there is no effective treatment. Cell therapy appears to be an option that is of growing interest in neural studies. The main aim of this study was to investigate the histological and physiological effects of transplantation the neuron-like cell (NLC)-derived mouse embryonic stem cells (mESCs) on the repair of brain lesions in an Alzheimer's animal model (AM) in rats. METHODS Behavioral experiments were conducted in the light hours in a Y-shaped maze device. Animals were randomly divided into five groups, with seven rats per group. The nucleus basalis of Meynert (NBM) was destroyed bilaterally with an electrical lesion (0.5 mA for 3 s). One week after the bilateral lesion of the NBM, the differentiated NLCs (0.1 mL) were injected with stereotaxic surgery using a Hamilton syringe at NBM coordinates, and behavioral and histological tests were performed by the Y-maze task and hematoxylin and eosin staining after five weeks of the lesion. Also, differentiated cells detected by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) analysis and fluorescent immunostaining. RESULTS The expression of neuronal markers including Nestin, Map2, NF-H, Tuj-1, GFAP and Olig-2 was surveyed by using the immunocytochemistry and qRT-PCR methods, and the results confirmed that the genes in question were expressed significantly more compared than the control sample. Five weeks after the cell transplantation in the AM, morphological and physiological investigation during the determination period confirmed improved disease state in the tested models. CONCLUSIONS It should be noted that by improving the neuronal connectivity in AM rat brains, the transplanted NLCs rescue Alzheimer's cognition. This research has presented some preclinical evidence that showed NLCs transplantation can be used for AM treatment.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Tayebeh Mohammadi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran; Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Ahmad Ali Moazedi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Azade Eskandary
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
24
|
El-Magd MA, Khalifa SF, A Alzahrani FA, Badawy AA, El-Shetry ES, Dawood LM, Alruwaili MM, Alrawaili HA, Risha EF, El-Taweel FM, Marei HE. Incensole acetate prevents beta-amyloid-induced neurotoxicity in human olfactory bulb neural stem cells. Biomed Pharmacother 2018; 105:813-823. [PMID: 29913410 DOI: 10.1016/j.biopha.2018.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/02/2018] [Accepted: 06/03/2018] [Indexed: 12/17/2022] Open
Abstract
β-Amyloid peptide (Aβ) is a potent neurotoxic protein associated with Alzheimer's disease (AD) which causes oxidative damage to neurons. Incensole acetate (IA) is a major constituent of Boswellia carterii resin, which has anti-inflammatory and protective properties against damage of a large verity of neural subtypes. However, this neuroprotective effect was not studied on human olfactory bulb neural stem cells (hOBNSCs). Herein, we evaluated this effect and studied the underlying mechanisms. Exposure to Aβ25-35 (5 and 10 μM for 24 h) inhibited proliferation (revealed by downregulation of Nestin and Sox2 gene expression), and induced differentiation (marked by increased expression of the immature neuronal marker Map2 and the astrocyte marker Gfap) of hOBNSCs. However, pre-treatment with IA (100 μM for 4 h) stimulated proliferation and differentiation of neuronal, rather than astrocyte, markers. Moreover, IA pretreatment significantly decreased the Aβ25-35-induced viability loss, apoptotic rate (revealed by decreased caspase 3 activity and protein expression, downregulated expression of Bax, caspase 8, cyto c, caspase3, and upregulated expression of Bcl2 mRNAs and proteins, in addition to elevated mitochondrial membrane potential and lowered intracellular Ca+2). IA reduced Aβ-mediated ROS production (revealed by decreased intracellular ROS and MDA level, and increased SOD, CAT, and GPX contents), and inhibited Aβ-induced inflammation (marked by down-regulated expression of IL1b, TNFa, NfKb, and Cox2 genes). IA also significantly upregulated mRNA and protein expression of Erk1/2 and Nrf2. Notably, IA increased the antioxidant enzyme heme oxygenase-1 (HO-1) expression and this effect was reversed by HO-1 inhibitor zinc protoporphyrin (ZnPP) leading to reduction of the neuroprotective effect of IA against Aβ-induced neurotoxicity. These findings clearly show the ability of IA to initiate proliferation and differentiation of neuronal progenitors in hOBNSCs and induce HO-1 expression, thereby protecting the hOBNSCs cells from Aβ25-35-induced oxidative cell death. Thus, IA may be applicable as a potential preventive agent for AD by its effect on hOBNSCs and could also be used as an adjuvant to hOBNSCs in cellular therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammed A El-Magd
- Department of Anatomy, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt.
| | - Sara F Khalifa
- Department of Chemistry, Faculty of Science, Damietta University, Egypt
| | - Faisal Abdulrahman A Alzahrani
- Department of Biological Sciences, Rabigh College of Science and Arts, King Abdulaziz University (Jeddah), Rabigh Branch, Rabigh 21911, Saudi Arabia
| | - Abdelnaser A Badawy
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Egypt
| | - Eman S El-Shetry
- Department of Human Anatomy, Faculty of Medicine, Zagazig University, Egypt
| | - Lamess M Dawood
- Department of Biochemistry, Faculty of Medicine, Tanta University, Egypt
| | - Mohammed M Alruwaili
- Medical Laboratory Technology Department, Faculty of Applied Medical Biosciences, Northern Border University, Arar City, Saudi Arabia
| | - Hedib A Alrawaili
- Medical Laboratory Sciences Department, School of Health Sciences, Quinnipiac University, Hamden, CT, USA
| | - Engi F Risha
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Fathy M El-Taweel
- Department of Chemistry, Faculty of Science, Damietta University, Egypt
| | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| |
Collapse
|
25
|
Lauzon MA, Faucheux N. A small peptide derived from BMP-9 can increase the effect of bFGF and NGF on SH-SY5Y cells differentiation. Mol Cell Neurosci 2018; 88:83-92. [PMID: 29341901 DOI: 10.1016/j.mcn.2018.01.003] [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: 09/14/2017] [Revised: 12/15/2017] [Accepted: 01/11/2018] [Indexed: 01/08/2023] Open
Abstract
The current aging of the world population will increase the number of people suffering from brain degenerative diseases such as Alzheimer's disease (AD). There are evidence showing that the use of growth factors such as BMP-9 could restored cognitive function as it acts on many AD hallmarks at the same time. However, BMP-9 is a big protein expensive to produce that can hardly access the central nervous system. We have therefore developed a small peptide, SpBMP-9, derived from the knuckle epitope of BMP-9 and showed its therapeutic potential in a previous study. Since it is known that the native protein, BMP-9, can act in synergy with other growth factors in the context of AD, here we study the potential synergistic effect of various combinations of SpBMP-9 with bFGF, EGF, IGF-2 or NGF on the cholinergic differentiation of human neuroblastoma cells SH-SY5Y. We found that, in opposition to IGF-2 or EGF, the combination of SpBMP-9 with bFGF or NGF can stimulate to a greater extent the neurite outgrowth and neuronal differentiation toward the cholinergic phenotype as shown by expression and localization of the neuronal markers NSE and VAchT and the staining of intracellular calcium. Those results strongly suggest that SpBMP-9 plus NGF or bFGF are promising therapeutic combinations against AD that required further attention.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada; Clinical Research Center of Centre Hospitalier Universitaire de Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada; Pharmacology Institute of Sherbrooke, 12e Avenue N, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
26
|
Song CG, Zhang YZ, Wu HN, Cao XL, Guo CJ, Li YQ, Zheng MH, Han H. Stem cells: a promising candidate to treat neurological disorders. Neural Regen Res 2018; 13:1294-1304. [PMID: 30028342 PMCID: PMC6065243 DOI: 10.4103/1673-5374.235085] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neurologic impairments are usually irreversible as a result of limited regeneration in the central nervous system. Therefore, based on the regenerative capacity of stem cells, transplantation therapies of various stem cells have been tested in basic research and preclinical trials, and some have shown great prospects. This manuscript overviews the cellular and molecular characteristics of embryonic stem cells, induced pluripotent stem cells, neural stem cells, retinal stem/progenitor cells, mesenchymal stem/stromal cells, and their derivatives in vivo and in vitro as sources for regenerative therapy. These cells have all been considered as candidates to treat several major neurological disorders and diseases, owing to their self-renewal capacity, multi-directional differentiation, neurotrophic properties, and immune modulation effects. We also review representative basic research and recent clinical trials using stem cells for neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, and age-related macular degeneration, as well as traumatic brain injury and glioblastoma. In spite of a few unsuccessful cases, risks of tumorigenicity, and ethical concerns, most results of animal experiments and clinical trials demonstrate efficacious therapeutic effects of stem cells in the treatment of nervous system disease. In summary, these emerging findings in regenerative medicine are likely to contribute to breakthroughs in the treatment of neurological disorders. Thus, stem cells are a promising candidate for the treatment of nervous system diseases.
Collapse
Affiliation(s)
- Chang-Geng Song
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yi-Zhe Zhang
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Hai-Ning Wu
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiu-Li Cao
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Chen-Jun Guo
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yong-Qiang Li
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Min-Hua Zheng
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Hua Han
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
27
|
Mukhamedyarov MA, Leushina AV, Tikhonova AE, Petukhova EO, Garanina EE, Ben Taleb R, Kaligin MS, Mukhamedshina YO, Rizvanov AA, Zefirov AL, Islamov RR. Intravenous Transplantation of Human Umbilical Cord Blood Mononuclear Cells Overexpressing Nerve Growth Factor Improves Spatial Memory in APP/PS1 Transgenic Mice with a Model of Alzheimer’s Disease. BIONANOSCIENCE 2017. [DOI: 10.1007/s12668-017-0497-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Marei HES, El-Gamal A, Althani A, Afifi N, Abd-Elmaksoud A, Farag A, Cenciarelli C, Thomas C, Anwarul H. Cholinergic and dopaminergic neuronal differentiation of human adipose tissue derived mesenchymal stem cells. J Cell Physiol 2017; 233:936-945. [PMID: 28369825 DOI: 10.1002/jcp.25937] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various cell types such as cartilage, bone, and fat cells. Recent studies have shown that induction of MSCs in vitro by growth factors including epidermal growth factor (EGF) and fibroblast growth factor (FGF2) causes them to differentiate into neural like cells. These cultures also express ChAT, a cholinergic marker; and TH, a dopaminergic marker for neural cells. To establish a protocol with maximum differentiation potential, we examined MSCs under three experimental culture conditions using neural induction media containing FGF2, EGF, BMP-9, retinoic acid, and heparin. Adipose-derived MSCs were extracted and expanded in vitro for 3 passages after reaching >80% confluency, for a total duration of 9 days. Cells were then characterized by flow cytometry for CD markers as CD44 positive and CD45 negative. MSCs were then treated with neural induction media and were characterized by morphological changes and Q-PCR. Differentiated MSCs expressed markers for immature and mature neurons; β Tubulin III (TUBB3) and MAP2, respectively, showing the neural potential of these cells to differentiate into functional neurons. Improved protocols for MSCs induction will facilitate and ensure the reproducibility and standard production of MSCs for therapeutic applications in neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Aya El-Gamal
- Faculty of Veterinary Medicine, Department of Cytology and Histology, Mansoura University, Mansoura, Egypt
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Ahmed Abd-Elmaksoud
- Faculty of Veterinary Medicine, Department of Cytology and Histology, Mansoura University, Mansoura, Egypt
| | - Amany Farag
- Faculty of Veterinary Medicine, Department of Cytology and Histology, Mansoura University, Mansoura, Egypt
| | | | - Caceci Thomas
- Department of Biomedical Sciences, Virginia Tech Carilion School of Medicine, Roanoke, Virginia
| | - Hasan Anwarul
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
29
|
Nanotechnological strategies for nerve growth factor delivery: Therapeutic implications in Alzheimer’s disease. Pharmacol Res 2017; 120:68-87. [DOI: 10.1016/j.phrs.2017.03.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 02/23/2017] [Accepted: 03/22/2017] [Indexed: 12/30/2022]
|
30
|
Marei HE, Elnegiry AA, Zaghloul A, Althani A, Afifi N, Abd-Elmaksoud A, Farag A, Lashen S, Rezk S, Shouman Z, Cenciarelli C, Hasan A. Nanotubes impregnated human olfactory bulb neural stem cells promote neuronal differentiation in Trimethyltin-induced neurodegeneration rat model. J Cell Physiol 2017; 232:3586-3597. [PMID: 28121007 DOI: 10.1002/jcp.25826] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 01/03/2017] [Accepted: 01/24/2017] [Indexed: 12/12/2022]
Abstract
Neural stem cells (NSCs) are multipotent self-renewing cells that could be used in cellular-based therapy for a wide variety of neurodegenerative diseases including Alzheimer's diseases (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Being multipotent in nature, they are practically capable of giving rise to major cell types of the nervous tissue including neurons, astrocytes, and oligodendrocytes. This is in marked contrast to neural progenitor cells which are committed to a specific lineage fate. In previous studies, we have demonstrated the ability of NSCs isolated from human olfactory bulb (OB) to survive, proliferate, differentiate, and restore cognitive and motor deficits associated with AD, and PD rat models, respectively. The use of carbon nanotubes (CNTs) to enhance the survivability and differentiation potential of NSCs following their in vivo engraftment have been recently suggested. Here, in order to assess the ability of CNTs to enhance the therapeutic potential of human OBNSCs for restoring cognitive deficits and neurodegenerative lesions, we co-engrafted CNTs and human OBNSCs in TMT-neurodegeneration rat model. The present study revealed that engrafted human OBNSCS-CNTs restored cognitive deficits, and neurodegenerative changes associated with TMT-induced rat neurodegeneration model. Moreover, the CNTs seemed to provide a support for engrafted OBNSCs, with increasing their tendency to differentiate into neurons rather than into glia cells. The present study indicate the marked ability of CNTs to enhance the therapeutic potential of human OBNSCs which qualify this novel therapeutic paradigm as a promising candidate for cell-based therapy of different neurodegenerative diseases.
Collapse
Affiliation(s)
- Hany E Marei
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Ahmed A Elnegiry
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Adel Zaghloul
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Ahmed Abd-Elmaksoud
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Amany Farag
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Samah Lashen
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Shymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Zeinab Shouman
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
31
|
Dunnett SB, Björklund A. Mechanisms and use of neural transplants for brain repair. PROGRESS IN BRAIN RESEARCH 2017; 230:1-51. [PMID: 28552225 DOI: 10.1016/bs.pbr.2016.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Under appropriate conditions, neural tissues transplanted into the adult mammalian brain can survive, integrate, and function so as to influence the behavior of the host, opening the prospect of repairing neuronal damage, and alleviating symptoms associated with neuronal injury or neurodegenerative disease. Alternative mechanisms of action have been postulated: nonspecific effects of surgery; neurotrophic and neuroprotective influences on disease progression and host plasticity; diffuse or locally regulated pharmacological delivery of deficient neurochemicals, neurotransmitters, or neurohormones; restitution of the neuronal and glial environment necessary for proper host neuronal support and processing; promoting local and long-distance host and graft axon growth; formation of reciprocal connections and reconstruction of local circuits within the host brain; and up to full integration and reconstruction of fully functional host neuronal networks. Analysis of neural transplants in a broad range of anatomical systems and disease models, on simple and complex classes of behavioral function and information processing, have indicated that all of these alternative mechanisms are likely to contribute in different circumstances. Thus, there is not a single or typical mode of graft function; rather grafts can and do function in multiple ways, specific to each particular context. Consequently, to develop an effective cell-based therapy, multiple dimensions must be considered: the target disease pathogenesis; the neurodegenerative basis of each type of physiological dysfunction or behavioral symptom; the nature of the repair required to alleviate or remediate the functional impairments of particular clinical relevance; and identification of a suitable cell source or delivery system, along with the site and method of implantation, that can achieve the sought for repair and recovery.
Collapse
|
32
|
Wang WW, Han JH, Wang L, Bao TH. Scutellarin may alleviate cognitive deficits in a mouse model of hypoxia by promoting proliferation and neuronal differentiation of neural stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:272-279. [PMID: 28392899 PMCID: PMC5378964 DOI: 10.22038/ijbms.2017.8355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective(s): Scutellarin, a flavonoid extracted from the medicinal herb Erigeron breviscapus Hand-Mazz, protects neurons from damage and inhibits glial activation. Here we examined whether scutellarin may also protect neurons from hypoxia-induced damage. Materials and Methods: Mice were exposed to hypoxia for 7 days and then administered scutellarin (50 mg/kg/d) or vehicle for 30 days Cognitive impairment in the two groups was assessed using the Morris water maze test, cell proliferation in the hippocampus was compared using 5-bromo-2-deoxyuridine (BrdU) immunohistochemistry, and hippocampal levels of nestin and neuronal class III β-tubulin (Tuj-1) were measured using Western blotting. These results were validated in vitro by treating cultured neural stem cells (NSCs) with scutellarin (30 μM). Results: Treating mice with scutellarin shortened escape times and increased the number of platform crossings, it increased the number of BrdU-positive proliferating cells in the hippocampus, and it up-regulated expression of nestin and Tuj-1. Treating NSC cultures with scutellarin increased the number of proliferating cells and the proportion of cells differentiating into neurons instead of astrocytes. The increase in NSC proliferation was associated with phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, while neuronal differentiation was associated with altered expression of differentiation-related genes. Conclusion: Scutellarin may alleviate cognitive impairment in a mouse model of hypoxia by promo-ting proliferation and neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Key Laboratory of Stem Cells and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan, PR China; Department of Anatomy and Development Biology, Monash University, Clayton, vic 3800, Australia
| | - Jian-Hong Han
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Tian-Hao Bao
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Mental Health Center of Kunming Medical University, Kunming City, Yunnan Province, PR China
| |
Collapse
|
33
|
Sensitive Tumorigenic Potential Evaluation of Adult Human Multipotent Neural Cells Immortalized by hTERT Gene Transduction. PLoS One 2016; 11:e0158639. [PMID: 27391353 PMCID: PMC4938125 DOI: 10.1371/journal.pone.0158639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 06/20/2016] [Indexed: 11/19/2022] Open
Abstract
Stem cells and therapeutic genes are emerging as a new therapeutic approach to treat various neurodegenerative diseases with few effective treatment options. However, potential formation of tumors by stem cells has hampered their clinical application. Moreover, adequate preclinical platforms to precisely test tumorigenic potential of stem cells are controversial. In this study, we compared the sensitivity of various animal models for in vivo stem cell tumorigenicity testing to identify the most sensitive platform. Then, tumorigenic potential of adult human multipotent neural cells (ahMNCs) immortalized by the human telomerase reverse transcriptase (hTERT) gene was examined as a stem cell model with therapeutic genes. When human glioblastoma (GBM) cells were injected into adult (4-6-week-old) Balb/c-nu, adult NOD/SCID, adult NOG, or neonate (1-2-week-old) NOG mice, the neonate NOG mice showed significantly faster tumorigenesis than that of the other groups regardless of intracranial or subcutaneous injection route. Two kinds of ahMNCs (682TL and 779TL) were primary cultured from surgical samples of patients with temporal lobe epilepsy. Although the ahMNCs were immortalized by lentiviral hTERT gene delivery (hTERT-682TL and hTERT-779TL), they did not form any detectable masses, even in the most sensitive neonate NOG mouse platform. Moreover, the hTERT-ahMNCs had no gross chromosomal abnormalities on a karyotype analysis. Taken together, our data suggest that neonate NOG mice could be a sensitive animal platform to test tumorigenic potential of stem cell therapeutics and that ahMNCs could be a genetically stable stem cell source with little tumorigenic activity to develop regenerative treatments for neurodegenerative diseases.
Collapse
|
34
|
Therapeutic potential of human olfactory bulb neural stem cells for spinal cord injury in rats. Spinal Cord 2016; 54:785-797. [PMID: 26882489 DOI: 10.1038/sc.2016.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/27/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
STUDY DESIGN Adult human olfactory bulb neural stem cells (OBNSCs) were isolated from human patients undergoing craniotomy for tumor resection. They were genetically engineered to overexpresses green fluorescent protein (GFP) to help trace them following engraftment. Spinal cord injury (SCI) was induced in rats using standard laminectomy protocol, and GFP-OBNSC were engrafted into rat model of SCI at day 7 post injury. Three rat groups were used: (i) Control group, (ii) Sham group (injected with cerebrospinal fluid) and treated group (engrafted with OBNSCs). Tissues from different groups were collected weekly up to 2 months. The collected tissues were fixed in 4% paraformaldehyde, processed for paraffin sectioning, immunohistochemically stained for different neuronal and glial markers and examined with bright-field fluorescent microscopy. Restoration of sensory motor functions we assessed on a weekly bases using the BBB score. OBJECTIVES To assess the therapeutic potential of OBNSCs-GFP and their ability to survive, proliferate, differentiate and to restore lost sensory motor functions following their engraftment in spinal cord injury (SCI). METHODS GFP-OBNSC were engrafted into a rat model of SCI at day 7 post injury and were followed-up to 8 weeks using behavioral and histochemical methods. RESULTS All transplanted animals exhibited successful engraftment. The survival rate was about 30% of initially transplanted cells. Twenty-seven percent of the engrafted cells differentiated along the NG2 and O4-positive oligodendrocyte lineage, 16% into MAP2 and β-tubulin-positive neurons, and 56% into GFAP-positive astrocytes. CONCLUSION GFP-OBNSCs had survived for >8 weeks after engraftment and were differentiated into neurons, astrocytes and oligodendrocytes, The engrafted cells were distributed throughout gray and white matter of the cord with no evidence of abnormal morphology or any mass formation indicative of tumorigenesis. However, the engrafted cells failed to restore lost sensory and motor functions as evident from behavioral analysis using the BBB score test.
Collapse
|
35
|
Ruzicka J, Kulijewicz-Nawrot M, Rodrigez-Arellano JJ, Jendelova P, Sykova E. Mesenchymal Stem Cells Preserve Working Memory in the 3xTg-AD Mouse Model of Alzheimer's Disease. Int J Mol Sci 2016; 17:ijms17020152. [PMID: 26821012 PMCID: PMC4783886 DOI: 10.3390/ijms17020152] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/12/2016] [Accepted: 01/20/2016] [Indexed: 12/26/2022] Open
Abstract
The transplantation of stem cells may have a therapeutic effect on the pathogenesis and progression of neurodegenerative disorders. In the present study, we transplanted human mesenchymal stem cells (MSCs) into the lateral ventricle of a triple transgenic mouse model of Alzheimer´s disease (3xTg-AD) at the age of eight months. We evaluated spatial reference and working memory after MSC treatment and the possible underlying mechanisms, such as the influence of transplanted MSCs on neurogenesis in the subventricular zone (SVZ) and the expression levels of a 56 kDa oligomer of amyloid β (Aβ*56), glutamine synthetase (GS) and glutamate transporters (Glutamate aspartate transporter (GLAST) and Glutamate transporter-1 (GLT-1)) in the entorhinal and prefrontal cortices and the hippocampus. At 14 months of age we observed the preservation of working memory in MSC-treated 3xTg-AD mice, suggesting that such preservation might be due to the protective effect of MSCs on GS levels and the considerable downregulation of Aβ*56 levels in the entorhinal cortex. These changes were observed six months after transplantation, accompanied by clusters of proliferating cells in the SVZ. Since the grafted cells did not survive for the whole experimental period, it is likely that the observed effects could have been transiently more pronounced at earlier time points than at six months after cell application.
Collapse
Affiliation(s)
- Jiri Ruzicka
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague 150 06, Czech Republic.
| | - Magdalena Kulijewicz-Nawrot
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
| | - Jose Julio Rodrigez-Arellano
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
- Functional Neuroanatomy Laboratory, Department of Neuroscience, Faculty of Medicine, the University of the Basque Country, 48940 Leioa, Spain.
| | - Pavla Jendelova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague 150 06, Czech Republic.
| | - Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague 142 20, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague 150 06, Czech Republic.
| |
Collapse
|
36
|
The Emerging Therapeutic Role of NGF in Alzheimer’s Disease. Neurochem Res 2016; 41:1211-8. [DOI: 10.1007/s11064-016-1829-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/08/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
|
37
|
Huang M, Hu M, Song Q, Song H, Huang J, Gu X, Wang X, Chen J, Kang T, Feng X, Jiang D, Zheng G, Chen H, Gao X. GM1-Modified Lipoprotein-like Nanoparticle: Multifunctional Nanoplatform for the Combination Therapy of Alzheimer's Disease. ACS NANO 2015; 9:10801-16. [PMID: 26440073 DOI: 10.1021/acsnano.5b03124] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Alzheimer's disease (AD) exerts a heavy health burden for modern society and has a complicated pathological background. The accumulation of extracellular β-amyloid (Aβ) is crucial in AD pathogenesis, and Aβ-initiated secondary pathological processes could independently lead to neuronal degeneration and pathogenesis in AD. Thus, the development of combination therapeutics that can not only accelerate Aβ clearance but also simultaneously protect neurons or inhibit other subsequent pathological cascade represents a promising strategy for AD intervention. Here, we designed a nanostructure, monosialotetrahexosylganglioside (GM1)-modified reconstituted high density lipoprotein (GM1-rHDL), that possesses antibody-like high binding affinity to Aβ, facilitates Aβ degradation by microglia, and Aβ efflux across the blood-brain barrier (BBB), displays high brain biodistribution efficiency following intranasal administration, and simultaneously allows the efficient loading of a neuroprotective peptide, NAP, as a nanoparticulate drug delivery system for the combination therapy of AD. The resulting multifunctional nanostructure, αNAP-GM1-rHDL, was found to be able to protect neurons from Aβ(1-42) oligomer/glutamic acid-induced cell toxicity better than GM1-rHDL in vitro and reduced Aβ deposition, ameliorated neurologic changes, and rescued memory loss more efficiently than both αNAP solution and GM1-rHDL in AD model mice following intranasal administration with no observable cytotoxicity noted. Taken together, this work presents direct experimental evidence of the rational design of a biomimetic nanostructure to serve as a safe and efficient multifunctional nanoplatform for the combination therapy of AD.
Collapse
Affiliation(s)
- Meng Huang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Meng Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Qingxiang Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Huahua Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Jialin Huang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Xiao Gu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Xiaolin Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Jun Chen
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Ting Kang
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Xingye Feng
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Di Jiang
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, School of Pharmacy, Fudan University , 826 Zhangheng Road, Shanghai 201203, People's Republic of China
| | - Gang Zheng
- Department of Medical Biophysics and Ontario Cancer Institute, University of Toronto , Toronto, Ontario M5G 1L7, Canada
| | - Hongzhuan Chen
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | - Xiaoling Gao
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine , 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| |
Collapse
|
38
|
Kanninen KM, Pomeshchik Y, Leinonen H, Malm T, Koistinaho J, Levonen AL. Applications of the Keap1-Nrf2 system for gene and cell therapy. Free Radic Biol Med 2015; 88:350-361. [PMID: 26164630 DOI: 10.1016/j.freeradbiomed.2015.06.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 01/15/2023]
Abstract
Oxidative stress has been implicated to play a role in a number of acute and chronic diseases including acute injuries of the central nervous system, neurodegenerative and cardiovascular diseases, and cancer. The redox-activated transcription factor Nrf2 has been shown to protect many different cell types and organs from a variety of toxic insults, whereas in many cancers, unchecked Nrf2 activity increases the expression of cytoprotective genes and, consequently, provides growth advantage to cancerous cells. Herein, we discuss current preclinical gene therapy approaches to either increase or decrease Nrf2 activity with a special reference to neurological diseases and cancer. In addition, we discuss the role of Nrf2 in stem cell therapy for neurological disorders.
Collapse
Affiliation(s)
- Katja M Kanninen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Yuriy Pomeshchik
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Hanna Leinonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Tarja Malm
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland.
| | - Anna-Liisa Levonen
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland.
| |
Collapse
|
39
|
Marei HES, Lashen S, Farag A, Althani A, Afifi N, A AE, Rezk S, Pallini R, Casalbore P, Cenciarelli C. Human olfactory bulb neural stem cells mitigate movement disorders in a rat model of Parkinson's disease. J Cell Physiol 2015; 230:1614-29. [PMID: 25536543 DOI: 10.1002/jcp.24909] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 12/18/2014] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by the loss of midbrain dopaminergic (DA) neurons. Neural stem cells (NSCs) are multipotent stem cells that are capable of differentiating into different neuronal and glial elements. The production of DA neurons from NSCs could potentially alleviate behavioral deficits in Parkinsonian patients; timely intervention with NSCs might provide a therapeutic strategy for PD. We have isolated and generated highly enriched cultures of neural stem/progenitor cells from the human olfactory bulb (OB). If NSCs can be obtained from OB, it would alleviate ethical concerns associated with the use of embryonic tissue, and provide an easily accessible cell source that would preclude the need for invasive brain surgery. Following isolation and culture, olfactory bulb neural stem cells (OBNSCs) were genetically engineered to express hNGF and GFP. The hNFG-GFP-OBNSCs were transplanted into the striatum of 6-hydroxydopamin (6-OHDA) Parkinsonian rats. The grafted cells survived in the lesion environment for more than eight weeks after implantation with no tumor formation. The grafted cells differentiated in vivo into oligodendrocyte-like (25 ± 2.88%), neuron-like (52.63 ± 4.16%), and astrocyte -like (22.36 ± 1.56%) lineages, which we differentiated based on morphological and immunohistochemical criteria. Transplanted rats exhibited a significant partial correction in stepping and placing in non-pharmacological behavioral tests, pole and rotarod tests. Taken together, our data encourage further investigations of the possible use of OBNSCs as a promising cell-based therapeutic strategy for Parkinson's disease.
Collapse
Affiliation(s)
- Hany E S Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | | | | | | | | | | | | | | | | |
Collapse
|