1
|
Stepien TL. An Approximate Bayesian Computation Approach for Embryonic Astrocyte Migration Model Reduction. Bull Math Biol 2024; 86:126. [PMID: 39269511 DOI: 10.1007/s11538-024-01354-5] [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: 02/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
During embryonic development of the retina of the eye, astrocytes, a type of glial cell, migrate over the retinal surface and form a dynamic mesh. This mesh then serves as scaffolding for blood vessels to form the retinal vasculature network that supplies oxygen and nutrients to the inner portion of the retina. Astrocyte spreading proceeds in a radially symmetric manner over the retinal surface. Additionally, astrocytes mature from astrocyte precursor cells (APCs) to immature perinatal astrocytes (IPAs) during this embryonic stage. We extend a previously-developed continuum model that describes tension-driven migration and oxygen and growth factor influenced proliferation and differentiation. Comparing numerical simulations to experimental data, we identify model equation components that can be removed via model reduction using approximate Bayesian computation (ABC). Our results verify experimental studies indicating that the choroid oxygen supply plays a negligible role in promoting differentiation of APCs into IPAs and in promoting IPA proliferation, and the hyaloid artery oxygen supply and APC apoptosis play negligible roles in astrocyte spreading and differentiation.
Collapse
Affiliation(s)
- Tracy L Stepien
- Department of Mathematics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
2
|
Mitchell CL, Kurouski D. Novel strategies in Parkinson's disease treatment: a review. Front Mol Neurosci 2024; 17:1431079. [PMID: 39183754 PMCID: PMC11341544 DOI: 10.3389/fnmol.2024.1431079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
An unprecedented extension of life expectancy observed during the past century drastically increased the number of patients diagnosed with Parkinson's diseases (PD) worldwide. Estimated costs of PD alone reached $52 billion per year, making effective neuroprotective treatments an urgent and unmet need. Current treatments of both AD and PD focus on mitigating the symptoms associated with these pathologies and are not neuroprotective. In this review, we discuss the most advanced therapeutic strategies that can be used to treat PD. We also critically review the shift of the therapeutic paradigm from a small molecule-based inhibition of protein aggregation to the utilization of natural degradation pathways and immune cells that are capable of degrading toxic amyloid deposits in the brain of PD patients.
Collapse
Affiliation(s)
- Charles L. Mitchell
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Dmitry Kurouski
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
3
|
Hastings N, Yu Y, Huang B, Middya S, Inaoka M, Erkamp NA, Mason RJ, Carnicer‐Lombarte A, Rahman S, Knowles TPJ, Bance M, Malliaras GG, Kotter MRN. Electrophysiological In Vitro Study of Long-Range Signal Transmission by Astrocytic Networks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301756. [PMID: 37485646 PMCID: PMC10582426 DOI: 10.1002/advs.202301756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Astrocytes are diverse brain cells that form large networks communicating via gap junctions and chemical transmitters. Despite recent advances, the functions of astrocytic networks in information processing in the brain are not fully understood. In culture, brain slices, and in vivo, astrocytes, and neurons grow in tight association, making it challenging to establish whether signals that spread within astrocytic networks communicate with neuronal groups at distant sites, or whether astrocytes solely respond to their local environments. A multi-electrode array (MEA)-based device called AstroMEA is designed to separate neuronal and astrocytic networks, thus allowing to study the transfer of chemical and/or electrical signals transmitted via astrocytic networks capable of changing neuronal electrical behavior. AstroMEA demonstrates that cortical astrocytic networks can induce a significant upregulation in the firing frequency of neurons in response to a theta-burst charge-balanced biphasic current stimulation (5 pulses of 100 Hz × 10 with 200 ms intervals, 2 s total duration) of a separate neuronal-astrocytic group in the absence of direct neuronal contact. This result corroborates the view of astrocytic networks as a parallel mechanism of signal transmission in the brain that is separate from the neuronal connectome. Translationally, it highlights the importance of astrocytic network protection as a treatment target.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Yi‐Lin Yu
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Department of Neurological SurgeryTri‐Service General HospitalNational Defence Medical CentreTaipei, Neihu District11490Taiwan
| | - Botian Huang
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - Sagnik Middya
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Misaki Inaoka
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Nadia A. Erkamp
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Roger J. Mason
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | | | - Saifur Rahman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of ChemistryCentre for Misfolding DiseasesUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
- Cavendish LaboratoryDepartment of PhysicsUniversity of CambridgeJ J Thomson AveCambridgeCB3 0HEUK
| | - Manohar Bance
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - George G. Malliaras
- Electrical Engineering DivisionDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Mark R. N. Kotter
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| |
Collapse
|
4
|
Wang T, Sun Y, Dettmer U. Astrocytes in Parkinson's Disease: From Role to Possible Intervention. Cells 2023; 12:2336. [PMID: 37830550 PMCID: PMC10572093 DOI: 10.3390/cells12192336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons. While neuronal dysfunction is central to PD, astrocytes also play important roles, both positive and negative, and such roles have not yet been fully explored. This literature review serves to highlight these roles and how the properties of astrocytes can be used to increase neuron survivability. Astrocytes normally have protective functions, such as releasing neurotrophic factors, metabolizing glutamate, transferring healthy mitochondria to neurons, or maintaining the blood-brain barrier. However, in PD, astrocytes can become dysfunctional and contribute to neurotoxicity, e.g., via impaired glutamate metabolism or the release of inflammatory cytokines. Therefore, astrocytes represent a double-edged sword. Restoring healthy astrocyte function and increasing the beneficial effects of astrocytes represents a promising therapeutic approach. Strategies such as promoting neurotrophin release, preventing harmful astrocyte reactivity, or utilizing regional astrocyte diversity may help restore neuroprotection.
Collapse
Affiliation(s)
- Tianyou Wang
- Collège Jean-de-Brébeuf, 3200 Chemin de la Côte-Sainte-Catherine, Montreal, QC H3T 1C1, Canada
| | - Yingqi Sun
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK;
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
5
|
Baranes K, Hastings N, Rahman S, Poulin N, Tavares JM, Kuan W, Syed N, Kunz M, Blighe K, Belgard TG, Kotter MRN. Transcription factor combinations that define human astrocyte identity encode significant variation of maturity and function. Glia 2023; 71:1870-1889. [PMID: 37029764 PMCID: PMC10952910 DOI: 10.1002/glia.24372] [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: 08/16/2021] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 04/09/2023]
Abstract
Increasing evidence indicates that cellular identity can be reduced to the distinct gene regulatory networks controlled by transcription factors (TFs). However, redundancy exists in these states as different combinations of TFs can induce broadly similar cell types. We previously demonstrated that by overcoming gene silencing, it is possible to deterministically reprogram human pluripotent stem cells directly into cell types of various lineages. In the present study we leverage the consistency and precision of our approach to explore four different TF combinations encoding astrocyte identity, based on previously published reports. Analysis of the resulting induced astrocytes (iAs) demonstrated that all four cassettes generate cells with the typical morphology of in vitro astrocytes, which expressed astrocyte-specific markers. The transcriptional profiles of all four iAs clustered tightly together and displayed similarities with mature human astrocytes, although maturity levels differed between cells. Importantly, we found that the TF cassettes induced iAs with distinct differences with regards to their cytokine response and calcium signaling. In vivo transplantation of selected iAs into immunocompromised rat brains demonstrated long term stability and integration. In conclusion, all four TF combinations were able to induce stable astrocyte-like cells that were morphologically similar but showed subtle differences with respect to their transcriptome. These subtle differences translated into distinct differences with regards to cell function, that could be related to maturation state and/or regional identity of the resulting cells. This insight opens an opportunity to precision-engineer cells to meet functional requirements, for example, in the context of therapeutic cell transplantation.
Collapse
Affiliation(s)
- Koby Baranes
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Nataly Hastings
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Saifur Rahman
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Noah Poulin
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Joana M. Tavares
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| | - Wei‐Li Kuan
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| | - Najeeb Syed
- The Bioinformatics CROSanfordFlorida32771USA
| | - Meik Kunz
- The Bioinformatics CROSanfordFlorida32771USA
| | | | | | - Mark R. N. Kotter
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
- Wellcome‐MRC Cambridge Stem Cell Institute, University of CambridgeCambridgeCB2 0AWUK
| |
Collapse
|
6
|
Wang Y, Xia Y, Kou L, Yin S, Chi X, Li J, Sun Y, Wu J, Zhou Q, Zou W, Jin Z, Huang J, Xiong N, Wang T. Astrocyte-to-neuron reprogramming and crosstalk in the treatment of Parkinson's disease. Neurobiol Dis 2023:106224. [PMID: 37433411 DOI: 10.1016/j.nbd.2023.106224] [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: 05/03/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Parkinson's disease (PD) is currently the fastest growing disabling neurological disorder worldwide, with motor and non-motor symptoms being its main clinical manifestations. The primary pathological features include a reduction in the number of dopaminergic neurons in the substantia nigra and decrease in dopamine levels in the nigrostriatal pathway. Existing treatments only alleviate clinical symptoms and do not stop disease progression; slowing down the loss of dopaminergic neurons and stimulating their regeneration are emerging therapies. Preclinical studies have demonstrated that transplantation of dopamine cells generated from human embryonic or induced pluripotent stem cells can restore the loss of dopamine. However, the application of cell transplantation is limited owing to ethical controversies and the restricted source of cells. Until recently, the reprogramming of astrocytes to replenish lost dopaminergic neurons has provided a promising alternative therapy for PD. In addition, repair of mitochondrial perturbations, clearance of damaged mitochondria in astrocytes, and control of astrocyte inflammation may be extensively neuroprotective and beneficial against chronic neuroinflammation in PD. Therefore, this review primarily focuses on the progress and remaining issues in astrocyte reprogramming using transcription factors (TFs) and miRNAs, as well as exploring possible new targets for treating PD by repairing astrocytic mitochondria and reducing astrocytic inflammation.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
7
|
Chierzi S, Kacerovsky JB, Fok AHK, Lahaie S, Shibi Rosen A, Farmer WT, Murai KK. Astrocytes Transplanted during Early Postnatal Development Integrate, Mature, and Survive Long Term in Mouse Cortex. J Neurosci 2023; 43:1509-1529. [PMID: 36669885 PMCID: PMC10008063 DOI: 10.1523/jneurosci.0544-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
Astrocytes have complex structural, molecular, and physiological properties and form specialized microenvironments that support circuit-specific functions in the CNS. To better understand how astrocytes acquire their unique features, we transplanted immature mouse cortical astrocytes into the developing cortex of male and female mice and assessed their integration, maturation, and survival. Within days, transplanted astrocytes developed morphologies and acquired territories and tiling behavior typical of cortical astrocytes. At 35-47 d post-transplantation, astrocytes appeared morphologically mature and expressed levels of EAAT2/GLT1 similar to nontransplanted astrocytes. Transplanted astrocytes also supported excitatory/inhibitory (E/I) presynaptic terminals within their territories, and displayed normal Ca2+ events. Transplanted astrocytes showed initially reduced expression of aquaporin 4 (AQP4) at endfeet and elevated expression of EAAT1/GLAST, with both proteins showing normalized expression by 110 d and one year post-transplantation, respectively. To understand how specific brain regions support astrocytic integration and maturation, we transplanted cortical astrocytes into the developing cerebellum. Cortical astrocytes interlaced with Bergmann glia (BG) in the cerebellar molecular layer to establish discrete territories. However, transplanted astrocytes retained many cortical astrocytic features including higher levels of EAAT2/GLT1, lower levels of EAAT1/GLAST, and the absence of expression of the AMPAR subunit GluA1. Collectively, our findings demonstrate that immature cortical astrocytes integrate, mature, and survive (more than one year) following transplantation and retain cortical astrocytic properties. Astrocytic transplantation can be useful for investigating cell-autonomous (intrinsic) and non-cell-autonomous (environmental) mechanisms contributing to astrocytic development/diversity, and for determining the optimal timing for transplanting astrocytes for cellular delivery or replacement in regenerative medicine.SIGNIFICANCE STATEMENT The mechanisms that enable astrocytes to acquire diverse molecular and structural properties remain to be better understood. In this study, we systematically analyzed the properties of cortical astrocytes following their transplantation to the early postnatal brain. We found that immature cortical astrocytes transplanted into cerebral cortex during early postnatal mouse development integrate and establish normal astrocytic properties, and show long-term survival in vivo (more than one year). In contrast, transplanted cortical astrocytes display reduced or altered ability to integrate into the more mature cerebral cortex or developing cerebellum, respectively. This study demonstrates the developmental potential of transplanted cortical astrocytes and provides an approach to tease apart cell-autonomous (intrinsic) and non-cell-autonomous (environmental) mechanisms that determine the structural, molecular, and physiological phenotype of astrocytes.
Collapse
Affiliation(s)
- Sabrina Chierzi
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - J Benjamin Kacerovsky
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - Albert H K Fok
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - Sylvie Lahaie
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - Arielle Shibi Rosen
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - W Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
- Quantitative Life Sciences Graduate Program, McGill University, Montreal, Quebec H3A 2A7, Canada
| |
Collapse
|
8
|
Yang Z, Gong M, Yang C, Chen C, Zhang K. Applications of Induced Pluripotent Stem Cell-Derived Glia in Brain Disease Research and Treatment. Handb Exp Pharmacol 2023; 281:103-140. [PMID: 37735301 DOI: 10.1007/164_2023_697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Glia are integral components of neural networks and are crucial in both physiological functions and pathological processes of the brain. Many brain diseases involve glial abnormalities, including inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. Induced pluripotent stem cell (iPSC)-derived glia provide opportunities to study the contributions of glia in human brain diseases. These cells have been used for human disease modeling as well as generating new therapies. This chapter introduces glial involvement in brain diseases, then summarizes different methods of generating iPSC-derived glia disease models of these cells. Finally, strategies for treating disease using iPSC-derived glia are discussed. The goal of this chapter is to provide an overview and shed light on the applications of iPSC-derived glia in brain disease research and treatment.
Collapse
Affiliation(s)
- Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chuanyan Yang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
9
|
Voronkov DN, Stavrovskaya AV, Guschina AS, Olshansky AS, Lebedeva OS, Eremeev AV, Lagarkova MA. Morphological Characterization of Astrocytes in a Xenograft of Human iPSCDerived Neural Precursor Cells. Acta Naturae 2022; 14:100-108. [PMID: 36348713 PMCID: PMC9611864 DOI: 10.32607/actanaturae.11710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/22/2022] [Indexed: 09/07/2024] Open
Abstract
Transplantation of a mixed astrocyte and neuron culture is of interest in the development of cell therapies for neurodegenerative diseases. In this case, an assessment of engraftment requires a detailed morphological characterization, in particular an analysis of the neuronal and glial populations. In the experiment performed, human iPSC-derived neural progenitors transplanted into a rat striatum produced a mixed neuron and astrocyte population in vivo by the sixth month after transplantation. The morphological characteristics and neurochemical profile of the xenografted astrocytes were similar to those of mature human astroglia. Unlike neurons, astrocytes migrated to the surrounding structures and the density and pattern of their distribution in the striatum and cerebral cortex differed, which indicates that the microenvironment affects human glia integration. The graft was characterized by the zonal features of glial cell morphology, which was a reflection of cell maturation in the central area, glial shaft formation around the transplanted neurons, and migration to the surrounding structures.
Collapse
Affiliation(s)
| | | | | | | | - O. S. Lebedeva
- Federal Research and Clinical Center of Physical Chemical Medicine of the Federal Medical and Biological Agency of the Russian Federation, Moscow, 119435 Russia
| | - A. V. Eremeev
- Federal Research and Clinical Center of Physical Chemical Medicine of the Federal Medical and Biological Agency of the Russian Federation, Moscow, 119435 Russia
| | - M. A. Lagarkova
- Federal Research and Clinical Center of Physical Chemical Medicine of the Federal Medical and Biological Agency of the Russian Federation, Moscow, 119435 Russia
| |
Collapse
|
10
|
The promise of the TGF-β superfamily as a therapeutic target for Parkinson's disease. Neurobiol Dis 2022; 171:105805. [PMID: 35764291 DOI: 10.1016/j.nbd.2022.105805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
A large body of evidence underscore the regulatory role of TGF-β superfamily in the central nervous system. Components of the TGF-β superfamily modulate key events during embryonic brain development and adult brain tissue injury repair. With respect to Parkinson's disease (PD), TGF-ß signaling pathways are implicated in the differentiation, maintenance and synaptic function of the dopaminergic neurons, as well as in processes related to the activation state of astrocytes and microglia. In vitro and in vivo studies using toxin models, have interrogated on the dopaminotrophic and protective role of the TGF-β superfamily members. The evolution of genetic and animal models of PD that more closely recapitulate the disease condition has made possible the dissection of intracellular pathways in response to TGF-ß treatment. Although the first clinical trials using GDNF did not meet their primary endpoints, substantial work has been carried out to reappraise the TGF-β superfamily's clinical benefit.
Collapse
|
11
|
Hastings N, Kuan WL, Osborne A, Kotter MRN. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant 2022; 31:9636897221105499. [PMID: 35770772 PMCID: PMC9251977 DOI: 10.1177/09636897221105499] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson's, Alzheimer's, and Huntington's diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Kurosaki T, Sakano H, Pröschel C, Wheeler J, Hewko A, Maquat LE. NMD abnormalities during brain development in the Fmr1-knockout mouse model of fragile X syndrome. Genome Biol 2021; 22:317. [PMID: 34784943 PMCID: PMC8597091 DOI: 10.1186/s13059-021-02530-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells. RESULTS We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development. CONCLUSIONS We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention.
Collapse
Affiliation(s)
- Tatsuaki Kurosaki
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Hitomi Sakano
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Christoph Pröschel
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Stem Cell and Regenerative Medicine Institute, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Jason Wheeler
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Alexander Hewko
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Lynne E. Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| |
Collapse
|
13
|
Voronkov D, Stavrovskaya A, Olshanskiy A, Guschina A, Khudoerkov R, Illarioshkin S. The Influence of Striatal Astrocyte Dysfunction on Locomotor Activity in Dopamine-depleted Rats. Basic Clin Neurosci 2021; 12:767-776. [PMID: 35693141 PMCID: PMC9168813 DOI: 10.32598/bcn.2021.1923.1] [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] [Received: 07/08/2019] [Revised: 10/23/2019] [Accepted: 06/24/2020] [Indexed: 11/22/2022] Open
Abstract
Introduction Astrocyte dysfunction is the common pathology failing astrocyte-neuron interaction in neurological diseases, including Parkinson's Disease (PD). The present study aimed to evaluate the impacts of astrocytic dysfunction caused by striatal injections of selective glial toxin L-Aminoadipic Acid (L-AA) on the rats' locomotor activity in normal conditions and under alpha-methyl-p-tyrosine depletion of catecholamines synthesis. Methods Thirty-three male Wistar rats were used in the experiments. Intrastriatal L-AA injections (100 μg) were performed into the right striatum. Alpha-methyl-p-tyrosine (a-MT, 100 mg/kg, inhibitor of tyrosine hydroxylase) was intraperitoneally injected for catecholamine depletion. The animals were divided into 5 groups, as follows: 1. L-AA treated (n=7), 2. L-AA+a-MT treated (n=5), 3. Sham-operated (n=7), 4. Sham+a-MT treated (n=5), 5. Intact control (n=9). For assessing motor function, open field and beam walking tests were used on the third day after the operation. Neuronal and astrocyte markers (glial fibrillary acidic protein, glutamine synthetase, tyrosine hydroxylase, & neuronal nuclear antigen) were examined in the striatum by immunohistochemistry. Results Administrating L-AA led to astrocytic degeneration in the striatum. No neuronal death and disruption of dopaminergic terminals were detected. L-AA and a-MT-treated animals' distance traveled was significantly (P=0.047) shorter than the Sham-operated group injected with a-MT. In the walking beam test, the number of unilateral paw slippings was significantly (P<0.01) higher in the L-AA-treated group than Sham-operated animals. Administrating a-MT alone and L-AA did not change rats' performance in walking beam tests. Conclusion Astrocyte ablation in dopamine depleted striatum resulted in reduced motor activity and asymmetrical gait disturbances. These findings demonstrated the role of astroglia in motor function regulation in the nigrostriatal system and suggest the possible association of glial dysfunction with motor dysfunction in PD. Highlights The local administration of gliotoxin L-aminoadipate in the striatum of rats causes astrocytic degeneration without affecting the neurons and nigrostriatal fibers.The failure of astrocyte-neuron coupling in the striatum leads to motor dysfunction such as gait disturbances and bradykinesia.The influence of astrocytic degeneration on behavior is preserved and enhanced in dopamine-depleted rats. Plain Language Summary Astrocytes are the nervous system's cells supporting the function of neurons. The failure of astrocyte-neuron interaction is observed in neurological diseases, including Parkinson's disease. We induced the aminoadipate-induced rat model of astrocytic dysfunction to evaluate the role of these cells in movement regulation. In our study, astrocytic dysfunction led to gait disturbances and impaired motor function. The results suggest a possible role of glial pathology in motor impairment in parkinsonism.
Collapse
|
14
|
Elahi A, Emerson J, Rudlong J, Keillor JW, Salois G, Visca A, Girardi P, Johnson GV, Pröschel C. Deletion or Inhibition of Astrocytic Transglutaminase 2 Promotes Functional Recovery after Spinal Cord Injury. Cells 2021; 10:2942. [PMID: 34831164 PMCID: PMC8616117 DOI: 10.3390/cells10112942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/23/2023] Open
Abstract
Following CNS injury, astrocytes become "reactive" and exhibit pro-regenerative or harmful properties. However, the molecular mechanisms that cause astrocytes to adopt either phenotype are not well understood. Transglutaminase 2 (TG2) plays a key role in regulating the response of astrocytes to insults. Here, we used mice in which TG2 was specifically deleted in astrocytes (Gfap-Cre+/- TG2fl/fl, referred to here as TG2-A-cKO) in a spinal cord contusion injury (SCI) model. Deletion of TG2 from astrocytes resulted in a significant improvement in motor function following SCI. GFAP and NG2 immunoreactivity, as well as number of SOX9 positive cells, were significantly reduced in TG2-A-cKO mice. RNA-seq analysis of spinal cords from TG2-A-cKO and control mice 3 days post-injury identified thirty-seven differentially expressed genes, all of which were increased in TG2-A-cKO mice. Pathway analysis revealed a prevalence for fatty acid metabolism, lipid storage and energy pathways, which play essential roles in neuron-astrocyte metabolic coupling. Excitingly, treatment of wild type mice with the selective TG2 inhibitor VA4 significantly improved functional recovery after SCI, similar to what was observed using the genetic model. These findings indicate the use of TG2 inhibitors as a novel strategy for the treatment of SCI and other CNS injuries.
Collapse
Affiliation(s)
- Anissa Elahi
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Jacen Emerson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Jacob Rudlong
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Garrick Salois
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Adam Visca
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| | - Peter Girardi
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Gail V.W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY 14620, USA; (J.E.); (J.R.); (P.G.)
| | - Christoph Pröschel
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14620, USA; (A.E.); (G.S.); (A.V.); (C.P.)
| |
Collapse
|
15
|
Valori CF, Possenti A, Brambilla L, Rossi D. Challenges and Opportunities of Targeting Astrocytes to Halt Neurodegenerative Disorders. Cells 2021; 10:cells10082019. [PMID: 34440788 PMCID: PMC8395029 DOI: 10.3390/cells10082019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders whose incidence is likely to duplicate in the next 30 years along with the progressive aging of the western population. Non-cell-specific therapeutics or therapeutics designed to tackle aberrant pathways within neurons failed to slow down or halt neurodegeneration. Yet, in the last few years, our knowledge of the importance of glial cells to maintain the central nervous system homeostasis in health conditions has increased exponentially, along with our awareness of their fundamental and multifaced role in pathological conditions. Among glial cells, astrocytes emerge as promising therapeutic targets in various neurodegenerative disorders. In this review, we present the latest evidence showing the astonishing level of specialization that astrocytes display to fulfill the demands of their neuronal partners as well as their plasticity upon injury. Then, we discuss the controversies that fuel the current debate on these cells. We tackle evidence of a potential beneficial effect of cell therapy, achieved by transplanting astrocytes or their precursors. Afterwards, we introduce the different strategies proposed to modulate astrocyte functions in neurodegeneration, ranging from lifestyle changes to environmental cues. Finally, we discuss the challenges and the recent advancements to develop astrocyte-specific delivery systems.
Collapse
Affiliation(s)
- Chiara F. Valori
- Molecular Neuropathology of Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Correspondence: (C.F.V.); (D.R.); Tel.: +49-7071-9254-122 (C.F.V.); +39-0382-592064 (D.R.)
| | - Agostino Possenti
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
| | - Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
- Correspondence: (C.F.V.); (D.R.); Tel.: +49-7071-9254-122 (C.F.V.); +39-0382-592064 (D.R.)
| |
Collapse
|
16
|
Hart CG, Karimi-Abdolrezaee S. Recent insights on astrocyte mechanisms in CNS homeostasis, pathology, and repair. J Neurosci Res 2021; 99:2427-2462. [PMID: 34259342 DOI: 10.1002/jnr.24922] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
Astrocytes play essential roles in development, homeostasis, injury, and repair of the central nervous system (CNS). Their development is tightly regulated by distinct spatial and temporal cues during embryogenesis and into adulthood throughout the CNS. Astrocytes have several important responsibilities such as regulating blood flow and permeability of the blood-CNS barrier, glucose metabolism and storage, synapse formation and function, and axon myelination. In CNS pathologies, astrocytes also play critical parts in both injury and repair mechanisms. Upon injury, they undergo a robust phenotypic shift known as "reactive astrogliosis," which results in both constructive and deleterious outcomes. Astrocyte activation and migration at the site of injury provides an early defense mechanism to minimize the extent of injury by enveloping the lesion area. However, astrogliosis also contributes to the inhibitory microenvironment of CNS injury and potentiate secondary injury mechanisms, such as inflammation, oxidative stress, and glutamate excitotoxicity, which facilitate neurodegeneration in CNS pathologies. Intriguingly, reactive astrocytes are increasingly a focus in current therapeutic strategies as their activation can be modulated toward a neuroprotective and reparative phenotype. This review will discuss recent advancements in knowledge regarding the development and role of astrocytes in the healthy and pathological CNS. We will also review how astrocytes have been genetically modified to optimize their reparative potential after injury, and how they may be transdifferentiated into neurons and oligodendrocytes to promote repair after CNS injury and neurodegeneration.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Ding ZB, Song LJ, Wang Q, Kumar G, Yan YQ, Ma CG. Astrocytes: a double-edged sword in neurodegenerative diseases. Neural Regen Res 2021; 16:1702-1710. [PMID: 33510058 PMCID: PMC8328766 DOI: 10.4103/1673-5374.306064] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play multifaceted and vital roles in maintaining neurophysiological function of the central nervous system by regulating homeostasis, increasing synaptic plasticity, and sustaining neuroprotective effects. Astrocytes become activated as a result of inflammatory responses during the progression of pathological changes associated with neurodegenerative disorders. Reactive astrocytes (neurotoxic A1 and neuroprotective A2) are triggered during disease progression and pathogenesis due to neuroinflammation and ischemia. However, only a limited body of literature describes morphological and functional changes of astrocytes during the progression of neurodegenerative diseases. The present review investigated the detrimental and beneficial roles of astrocytes in neurodegenerative diseases reported in recent studies, as these cells have promising therapeutic potential and offer new approaches for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Yu-Qing Yan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| |
Collapse
|
18
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
19
|
Hart CG, Karimi-Abdolrezaee S. Bone morphogenetic proteins: New insights into their roles and mechanisms in CNS development, pathology and repair. Exp Neurol 2020; 334:113455. [PMID: 32877654 DOI: 10.1016/j.expneurol.2020.113455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are a highly conserved and diverse family of proteins that play essential roles in various stages of development including the formation and patterning of the central nervous system (CNS). Bioavailability and function of BMPs are regulated by input from a plethora of transcription factors and signaling pathways. Intriguingly, recent literature has uncovered novel roles for BMPs in regulating homeostatic and pathological responses in the adult CNS. Basal levels of BMP ligands and receptors are widely expressed in the adult brain and spinal cord with differential expression patterns across CNS regions, cell types and subcellular locations. Recent evidence indicates that several BMP isoforms are transiently or chronically upregulated in the aged or pathological CNS. Genetic knockout and pharmacological studies have elucidated that BMPs regulate several aspects of CNS injury and repair including cell survival and differentiation, reactive astrogliosis and glial scar formation, axon regeneration, and myelin preservation and repair. Several BMP isoforms can be upregulated in the injured or diseased CNS simultaneously yet exert complementary or opposing effects on the endogenous cell responses after injury. Emerging studies also show that dysregulation of BMPs is associated with various CNS pathologies. Interestingly, modulation of BMPs can lead to beneficial or detrimental effects on CNS injury and repair mechanisms in a ligand, temporally or spatially specific manner, which reflect the complexity of BMP signaling. Given the significance of BMPs in neurodevelopment, a better understanding of their role in the context of injury may provide new therapeutic targets for the pathologic CNS. This review will provide a timely overview on the foundation and recent advancements in knowledge regarding the role and mechanisms of BMP signaling in the developing and adult CNS, and their implications in pathological responses and repair processes after injury or diseases.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
20
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
21
|
Shahlaei M, Asl SM, Saeidifar M. Nanotechnology in gene delivery for neural regenerative medicine. NEURAL REGENERATIVE NANOMEDICINE 2020:123-157. [DOI: 10.1016/b978-0-12-820223-4.00005-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Astrocytes: Emerging Therapeutic Targets in Neurological Disorders. Trends Mol Med 2019; 25:750-759. [DOI: 10.1016/j.molmed.2019.04.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022]
|
23
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
24
|
Oksanen M, Lehtonen S, Jaronen M, Goldsteins G, Hämäläinen RH, Koistinaho J. Astrocyte alterations in neurodegenerative pathologies and their modeling in human induced pluripotent stem cell platforms. Cell Mol Life Sci 2019; 76:2739-2760. [PMID: 31016348 PMCID: PMC6588647 DOI: 10.1007/s00018-019-03111-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/06/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022]
Abstract
Astrocytes are the most abundant cell type in the brain. They were long considered only as passive support for neuronal cells. However, recent data have revealed many active roles for these cells both in maintenance of the normal physiological homeostasis in the brain as well as in neurodegeneration and disease. Moreover, human astrocytes have been found to be much more complex than their rodent counterparts, and to date, astrocytes are known to actively participate in a multitude of processes such as neurotransmitter uptake and recycling, gliotransmitter release, neuroenergetics, inflammation, modulation of synaptic activity, ionic balance, maintenance of the blood-brain barrier, and many other crucial functions of the brain. This review focuses on the role of astrocytes in human neurodegenerative disease and the potential of the novel stem cell-based platforms in modeling astrocytic functions in health and in disease.
Collapse
Affiliation(s)
- Minna Oksanen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Sarka Lehtonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, PO. Box 63, 00290, Helsinki, Finland
| | - Merja Jaronen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Gundars Goldsteins
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Riikka H Hämäläinen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland
| | - Jari Koistinaho
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210, Kuopio, Finland.
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, PO. Box 63, 00290, Helsinki, Finland.
| |
Collapse
|
25
|
Cell reprogramming approaches in gene- and cell-based therapies for Parkinson's disease. J Control Release 2018; 286:114-124. [PMID: 30026082 DOI: 10.1016/j.jconrel.2018.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.
Collapse
|
26
|
Growth Factors and Neuroglobin in Astrocyte Protection Against Neurodegeneration and Oxidative Stress. Mol Neurobiol 2018; 56:2339-2351. [PMID: 29982985 DOI: 10.1007/s12035-018-1203-9] [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: 04/10/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases, such as Parkinson and Alzheimer, are among the main public health issues in the world due to their effects on life quality and high mortality rates. Although neuronal death is the main cause of disruption in the central nervous system (CNS) elicited by these pathologies, other cells such as astrocytes are also affected. There is no treatment for preventing the cellular death during neurodegenerative processes, and current drug therapy is focused on decreasing the associated motor symptoms. For these reasons, it has been necessary to seek new therapeutical procedures, including the use of growth factors to reduce α-synuclein toxicity and misfolding in order to recover neuronal cells and astrocytes. Additionally, it has been shown that some growth factors are able to reduce the overproduction of reactive oxygen species (ROS), which are associated with neuronal death through activation of antioxidative enzymes such as catalase, superoxide dismutase, glutathione peroxidase, and neuroglobin. In the present review, we discuss the use of growth factors such as PDGF-BB, VEGF, BDNF, and the antioxidative enzyme neuroglobin in the protection of astrocytes and neurons during the development of neurodegenerative diseases.
Collapse
|
27
|
Loera-Valencia R, Piras A, Ismail MAM, Manchanda S, Eyjolfsdottir H, Saido TC, Johansson J, Eriksdotter M, Winblad B, Nilsson P. Targeting Alzheimer's disease with gene and cell therapies. J Intern Med 2018; 284:2-36. [PMID: 29582495 DOI: 10.1111/joim.12759] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) causes dementia in both young and old people affecting more than 40 million people worldwide. The two neuropathological hallmarks of the disease, amyloid beta (Aβ) plaques and neurofibrillary tangles consisting of protein tau are considered the major contributors to the disease. However, a more complete picture reveals significant neurodegeneration and decreased cell survival, neuroinflammation, changes in protein and energy homeostasis and alterations in lipid and cholesterol metabolism. In addition, gene and cell therapies for severe neurodegenerative disorders have recently improved technically in terms of safety and efficiency and have translated to the clinic showing encouraging results. Here, we review broadly current data within the field for potential targets that could modify AD through gene and cell therapy strategies. We envision that not only Aβ will be targeted in a disease-modifying treatment strategy but rather that a combination of treatments, possibly at different intervention times may prove beneficial in curing this devastating disease. These include decreased tau pathology, neuronal growth factors to support neurons and modulation of neuroinflammation for an appropriate immune response. Furthermore, cell based therapies may represent potential strategies in the future.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - A Piras
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M A M Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Neuro, Diseases of the Nervous System Patient Flow, Karolinska University Hospital, Huddinge, Sweden
| | - S Manchanda
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - H Eyjolfsdottir
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - T C Saido
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - J Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - P Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
28
|
Song JJ, Oh SM, Kwon OC, Wulansari N, Lee HS, Chang MY, Lee E, Sun W, Lee SE, Chang S, An H, Lee CJ, Lee SH. Cografting astrocytes improves cell therapeutic outcomes in a Parkinson's disease model. J Clin Invest 2017; 128:463-482. [PMID: 29227284 DOI: 10.1172/jci93924] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Transplantation of neural progenitor cells (NPCs) is a potential therapy for treating neurodegenerative disorders, but this approach has faced many challenges and limited success, primarily because of inhospitable host brain environments that interfere with enriched neuron engraftment and function. Astrocytes play neurotrophic roles in the developing and adult brain, making them potential candidates for helping with modification of hostile brain environments. In this study, we examined whether astrocytic function could be utilized to overcome the current limitations of cell-based therapies in a murine model of Parkinson's disease (PD) that is characterized by dopamine (DA) neuron degeneration in the midbrain. We show here that cografting astrocytes, especially those derived from the midbrain, remarkably enhanced NPC-based cell therapeutic outcomes along with robust DA neuron engraftment in PD rats for at least 6 months after transplantation. We further show that engineering of donor astrocytes with Nurr1 and Foxa2, transcription factors that were recently reported to polarize harmful immunogenic glia into the neuroprotective form, further promoted the neurotrophic actions of grafted astrocytes in the cell therapeutic approach. Collectively, these findings suggest that cografting astrocytes could be a potential strategy for successful cell therapeutic outcomes in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jae-Jin Song
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Sang-Min Oh
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Oh-Chan Kwon
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Noviana Wulansari
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Hyun-Seob Lee
- Genomic Core Facility, Transdisciplinary Research and Collaboration Division, Translational Research Institute, and.,Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and
| | - Eunsoo Lee
- Department of Anatomy and Division of Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Woong Sun
- Department of Anatomy and Division of Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Sang-Eun Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Heeyoung An
- Center for Neuroscience and.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, South Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - C Justin Lee
- Center for Neuroscience and.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine.,Hanyang Biomedical Research Institute, and.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| |
Collapse
|
29
|
Systemic Neutrophil Depletion Modulates the Migration and Fate of Transplanted Human Neural Stem Cells to Rescue Functional Repair. J Neurosci 2017; 37:9269-9287. [PMID: 28847814 DOI: 10.1523/jneurosci.2785-16.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 06/15/2017] [Accepted: 07/30/2017] [Indexed: 01/19/2023] Open
Abstract
The interaction of transplanted stem cells with local cellular and molecular cues in the host CNS microenvironment may affect the potential for repair by therapeutic cell populations. In this regard, spinal cord injury (SCI), Alzheimer's disease, and other neurological injuries and diseases all exhibit dramatic and dynamic changes to the host microenvironment over time. Previously, we reported that delayed transplantation of human CNS-derived neural stem cells (hCNS-SCns) at 9 or 30 d post-SCI (dpi) resulted in extensive donor cell migration, predominantly neuronal and oligodendrocytic donor cell differentiation, and functional locomotor improvements. Here, we report that acute transplantation of hCNS-SCns at 0 dpi resulted in localized astroglial differentiation of donor cells near the lesion epicenter and failure to produce functional improvement in an all-female immunodeficient mouse model. Critically, specific immunodepletion of neutrophils (polymorphonuclear leukocytes) blocked hCNS-SCns astroglial differentiation near the lesion epicenter and rescued the capacity of these cells to restore function. These data represent novel evidence that a host immune cell population can block the potential for functional repair derived from a therapeutic donor cell population, and support targeting the inflammatory microenvironment in combination with cell transplantation after SCI.SIGNIFICANCE STATEMENT The interaction of transplanted cells with local cellular and molecular cues in the host microenvironment is a key variable that may shape the translation of neurotransplantation research to the clinical spinal cord injury (SCI) human population, and few studies have investigated these events. We show that the specific immunodepletion of polymorphonuclear leukocyte neutrophils using anti-Ly6G inhibits donor cell astrogliosis and rescues the capacity of a donor cell population to promote locomotor improvement after SCI. Critically, our data demonstrate novel evidence that a specific host immune cell population can block the potential for functional repair derived from a therapeutic donor cell population.
Collapse
|
30
|
Can Astrocytes Be a Target for Precision Medicine? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:111-128. [DOI: 10.1007/978-3-319-60733-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
VEGF treatment during status epilepticus attenuates long-term seizure-associated alterations in astrocyte morphology. Epilepsy Behav 2017; 70:33-44. [PMID: 28410463 DOI: 10.1016/j.yebeh.2017.02.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/13/2017] [Indexed: 11/22/2022]
Abstract
Vascular endothelial growth factor (VEGF) treatment during pilocarpine-induced status epilepticus (SE) causes sustained preservation of behavioral function in rats in the absence of enduring neuroprotection (Nicoletti et al., 2010), suggesting the possibility that other cells or mechanisms could be involved in the beneficial effects of VEGF during SE. Astrocytes have been reported to contribute to epileptiform discharges in the hippocampus (Tian et al., 2005; Kang et al., 1998) and to express VEGF receptors (Krum & Rosenstein, 2002). We report here that VEGF treatment significantly alters multiple astrocyte parameters. This study investigated astrocyte morphology one month after SE in animals treated with VEGF or inactivated VEGF control protein during SE. Individual GFAP-immunostained astrocytes from CA1 and dentate gyrus hilus were traced and morphologically quantified, and both somatic and process structures were analyzed. VEGF treatment during SE significantly prevented post-SE increases in number of branch intersections, process length, and node count. Furthermore, analysis of distance to nearest neighboring astrocytes revealed that VEGF treatment significantly increased inter-astrocyte distance. Overall, VEGF treatment during SE did not significantly alter the shape of the astrocytes, but did prevent SE-induced changes in branching complexity, size, and spatial patterning. Because astrocyte morphology may be related to astrocyte function, it is possible that VEGF's enduring effects on astrocyte morphology may impact the functioning of the post-seizure hippocampus.
Collapse
|
32
|
Gowing G, Svendsen S, Svendsen CN. Ex vivo gene therapy for the treatment of neurological disorders. PROGRESS IN BRAIN RESEARCH 2017; 230:99-132. [PMID: 28552237 DOI: 10.1016/bs.pbr.2016.11.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ex vivo gene therapy involves the genetic modification of cells outside of the body to produce therapeutic factors and their subsequent transplantation back into patients. Various cell types can be genetically engineered. However, with the explosion in stem cell technologies, neural stem/progenitor cells and mesenchymal stem cells are most often used. The synergy between the effect of the new cell and the additional engineered properties can often provide significant benefits to neurodegenerative changes in the brain. In this review, we cover both preclinical animal studies and clinical human trials that have used ex vivo gene therapy to treat neurological disorders with a focus on Parkinson's disease, Huntington's disease, Alzheimer's disease, ALS, and stroke. We highlight some of the major advances in this field including new autologous sources of pluripotent stem cells, safer ways to introduce therapeutic transgenes, and various methods of gene regulation. We also address some of the remaining hurdles including tunable gene regulation, in vivo cell tracking, and rigorous experimental design. Overall, given the current outcomes from researchers and clinical trials, along with exciting new developments in ex vivo gene and cell therapy, we anticipate that successful treatments for neurological diseases will arise in the near future.
Collapse
Affiliation(s)
- Genevieve Gowing
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Soshana Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
33
|
Epilepsy-causing sequence variations in SIK1 disrupt synaptic activity response gene expression and affect neuronal morphology. Eur J Hum Genet 2016; 25:216-221. [PMID: 27966542 DOI: 10.1038/ejhg.2016.145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 12/30/2022] Open
Abstract
SIK1 syndrome is a newly described developmental epilepsy disorder caused by heterozygous mutations in the salt-inducible kinase SIK1. To better understand the pathophysiology of SIK1 syndrome, we studied the effects of SIK1 pathogenic sequence variations in human neurons. Primary human fetal cortical neurons were transfected with a lentiviral vector to overexpress wild-type and mutant SIK1 protein. We evaluated the transcriptional activity of known downstream gene targets in neurons expressing mutant SIK1 compared with wild type. We then assayed neuronal morphology by measuring neurite length, number and branching. Truncating SIK1 sequence variations were associated with abnormal MEF2C transcriptional activity and decreased MEF2C protein levels. Epilepsy-causing SIK1 sequence variations were associated with significantly decreased expression of ARC (activity-regulated cytoskeletal-associated) and other synaptic activity response element genes. Assay of mRNA levels for other MEF2C target genes NR4A1 (Nur77) and NRG1, found significantly, decreased the expression of these genes as well. The missense p.(Pro287Thr) SIK1 sequence variation was associated with abnormal neuronal morphology, with significant decreases in mean neurite length, mean number of neurites and a significant increase in proximal branches compared with wild type. Epilepsy-causing SIK1 sequence variations resulted in abnormalities in the MEF2C-ARC pathway of neuronal development and synapse activity response. This work provides the first insights into the mechanisms of pathogenesis in SIK1 syndrome, and extends the ARX-MEF2C pathway in the pathogenesis of developmental epilepsy.
Collapse
|
34
|
Role of Matricellular Proteins in Disorders of the Central Nervous System. Neurochem Res 2016; 42:858-875. [DOI: 10.1007/s11064-016-2088-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022]
|
35
|
Miyazaki I, Asanuma M. Serotonin 1A Receptors on Astrocytes as a Potential Target for the Treatment of Parkinson's Disease. Curr Med Chem 2016; 23:686-700. [PMID: 26795196 PMCID: PMC4997990 DOI: 10.2174/0929867323666160122115057] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/12/2015] [Accepted: 01/22/2016] [Indexed: 12/30/2022]
Abstract
Astrocytes are the most abundant neuron-supporting glial cells in the central nervous system. The neuroprotective role of astrocytes has been demonstrated in various neurological disorders such as amyotrophic lateral sclerosis, spinal cord injury, stroke and Parkinson’s disease (PD). Astrocyte dysfunction or loss-of-astrocytes increases the susceptibility of neurons to cell death, while astrocyte transplantation in animal studies has therapeutic advantage. We reported recently that stimulation of serotonin 1A (5-HT1A) receptors on astrocytes promoted astrocyte proliferation and upregulated antioxidative molecules to act as a neuroprotectant in parkinsonian mice. PD is a progressive neurodegenerative disease with motor symptoms such as tremor, bradykinesia, rigidity and postural instability, that are based on selective loss of nigrostriatal dopaminergic neurons, and with non-motor symptoms such as orthostatic hypotension and constipation based on peripheral neurodegeneration. Although dopaminergic therapy for managing the motor disability associated with PD is being assessed at present, the main challenge remains the development of neuroprotective or disease-modifying treatments. Therefore, it is desirable to find treatments that can reduce the progression of dopaminergic cell death. In this article, we summarize first the neuroprotective properties of astrocytes targeting certain molecules related to PD. Next, we review neuroprotective effects induced by stimulation of 5-HT1A receptors on astrocytes. The review discusses new promising therapeutic strategies based on neuroprotection against oxidative stress and prevention of dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| | | |
Collapse
|
36
|
Verkhratsky A, Steardo L, Parpura V, Montana V. Translational potential of astrocytes in brain disorders. Prog Neurobiol 2016; 144:188-205. [PMID: 26386136 PMCID: PMC4794425 DOI: 10.1016/j.pneurobio.2015.09.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/03/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
Fundamentally, all brain disorders can be broadly defined as the homeostatic failure of this organ. As the brain is composed of many different cells types, including but not limited to neurons and glia, it is only logical that all the cell types/constituents could play a role in health and disease. Yet, for a long time the sole conceptualization of brain pathology was focused on the well-being of neurons. Here, we challenge this neuron-centric view and present neuroglia as a key element in neuropathology, a process that has a toll on astrocytes, which undergo complex morpho-functional changes that can in turn affect the course of the disorder. Such changes can be grossly identified as reactivity, atrophy with loss of function and pathological remodeling. We outline the pathogenic potential of astrocytes in variety of disorders, ranging from neurotrauma, infection, toxic damage, stroke, epilepsy, neurodevelopmental, neurodegenerative and psychiatric disorders, Alexander disease to neoplastic changes seen in gliomas. We hope that in near future we would witness glial-based translational medicine with generation of deliverables for the containment and cure of disorders. We point out that such as a task will require a holistic and multi-disciplinary approach that will take in consideration the concerted operation of all the cell types in the brain.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Luca Steardo
- Department of Psychiatry, University of Naples, SUN, Largo Madonna delle Grazie, Naples, Italy
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine and Atomic Force Microscopy & Nanotechnology Laboratories, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vedrana Montana
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
37
|
Campbell A, Bushman J, Munger J, Noble M, Pröoschel C, Mayer-Pröoschel M. Mutation of ataxia-telangiectasia mutated is associated with dysfunctional glutathione homeostasis in cerebellar astroglia. Glia 2016; 64:227-39. [PMID: 26469940 PMCID: PMC5580048 DOI: 10.1002/glia.22925] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 09/10/2015] [Accepted: 09/14/2015] [Indexed: 11/11/2022]
Abstract
Astroglial dysfunction plays an important role in neurodegenerative diseases otherwise attributed to neuronal loss of function. Here we focus on the role of astroglia in ataxia-telangiectasia (A-T), a disease caused by mutations in the ataxia-telangiectasia mutated (ATM) gene. A hallmark of A-T pathology is progressive loss of cerebellar neurons, but the mechanisms that impact neuronal survival are unclear. We now provide a possible mechanism by which A-T astroglia affect the survival of cerebellar neurons. As astroglial functions are difficult to study in an in vivo setting, particularly in the cerebellum where these cells are intertwined with the far more numerous neurons, we conducted in vitro coculture experiments that allow for the generation and pharmacological manipulation of purified cell populations. Our analyses revealed that cerebellar astroglia isolated from Atm mutant mice show decreased expression of the cystine/glutamate exchanger subunit xCT, glutathione (GSH) reductase, and glutathione-S-transferase. We also found decreased levels of intercellular and secreted GSH in A-T astroglia. Metabolic labeling of l-cystine, the major precursor for GSH, revealed that a key component of the defect in A-T astroglia is an impaired ability to import this rate-limiting precursor for the production of GSH. This impairment resulted in suboptimal extracellular GSH supply, which in turn impaired survival of cerebellar neurons. We show that by circumventing the xCT-dependent import of L-cystine through addition of N-acetyl-L-cysteine (NAC) as an alternative cysteine source, we were able to restore GSH levels in A-T mutant astroglia providing a possible future avenue for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Andrew Campbell
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, 14642
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, New York, 14642
| | - Jared Bushman
- School of Pharmacy Health Sciences Center, University of Wyoming, Laramie, Wyoming, 82071
| | - Joshua Munger
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, New York, 14642
| | - Mark Noble
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, 14642
| | - Christoph Pröoschel
- Department of Biomedical Genetics, University of Rochester, Rochester, New York, 14642
| | | |
Collapse
|
38
|
Novel Approaches in Astrocyte Protection: from Experimental Methods to Computational Approaches. J Mol Neurosci 2016; 58:483-92. [DOI: 10.1007/s12031-016-0719-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/13/2016] [Indexed: 12/21/2022]
|
39
|
Rapid and robust generation of long-term self-renewing human neural stem cells with the ability to generate mature astroglia. Sci Rep 2015; 5:16321. [PMID: 26541394 PMCID: PMC4635383 DOI: 10.1038/srep16321] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cell bear the potential to differentiate into any desired cell type and hold large promise for disease-in-a-dish cell-modeling approaches. With the latest advances in the field of reprogramming technology, the generation of patient-specific cells has become a standard technology. However, directed and homogenous differentiation of human pluripotent stem cells into desired specific cell types remains an experimental challenge. Here, we report the development of a novel hiPSCs-based protocol enabling the generation of expandable homogenous human neural stem cells (hNSCs) that can be maintained under self-renewing conditions over high passage numbers. Our newly generated hNSCs retained differentiation potential as evidenced by the reliable generation of mature astrocytes that display typical properties as glutamate up-take and expression of aquaporin-4. The hNSC-derived astrocytes showed high activity of pyruvate carboxylase as assessed by stable isotope assisted metabolic profiling. Moreover, using a cell transplantation approach, we showed that grafted hNSCs were not only able to survive but also to differentiate into astroglial in vivo. Engraftments of pluripotent stem cells derived from somatic cells carry an inherent tumor formation potential. Our results demonstrate that hNSCs with self-renewing and differentiation potential may provide a safer alternative strategy, with promising applications especially for neurodegenerative disorders.
Collapse
|
40
|
Chen C, Chan A, Wen H, Chung SH, Deng W, Jiang P. Stem and Progenitor Cell-Derived Astroglia Therapies for Neurological Diseases. Trends Mol Med 2015; 21:715-729. [PMID: 26443123 DOI: 10.1016/j.molmed.2015.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023]
Abstract
Astroglia are a major cellular constituent of the central nervous system (CNS) and play crucial roles in brain development, function, and integrity. Increasing evidence demonstrates that astroglia dysfunction occurs in a variety of neurological disorders ranging from CNS injuries to genetic diseases and chronic degenerative conditions. These new insights herald the concept that transplantation of astroglia could be of therapeutic value in treating the injured or diseased CNS. Recent technological advances in the generation of human astroglia from stem and progenitor cells have been prominent. We propose that a better understanding of the suitability of astroglial cells in transplantation as well as of their therapeutic effects in animal models may lead to the establishment of astroglia-based therapies to treat neurological diseases.
Collapse
Affiliation(s)
- Chen Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Albert Chan
- Department of Pediatrics, University of California, Davis, CA, USA
| | - Han Wen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | | | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA.
| | - Peng Jiang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA; Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, NE, USA.
| |
Collapse
|
41
|
Abstract
Stem cell-based therapies hold considerable promise for many currently devastating neurological disorders. Substantial progress has been made in the derivation of disease-relevant human donor cell populations. Behavioral data in relevant animal models of disease have demonstrated therapeutic efficacy for several cell-based approaches. Consequently, cGMP grade cell products are currently being developed for first in human clinical trials in select disorders. Despite the therapeutic promise, the presumed mechanism of action of donor cell populations often remains insufficiently validated. It depends greatly on the properties of the transplanted cell type and the underlying host pathology. Several new technologies have become available to probe mechanisms of action in real time and to manipulate in vivo cell function and integration to enhance therapeutic efficacy. Results from such studies generate crucial insight into the nature of brain repair that can be achieved today and push the boundaries of what may be possible in the future.
Collapse
|
42
|
d'Anglemont de Tassigny X, Pascual A, López-Barneo J. GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015; 9:10. [PMID: 25762899 PMCID: PMC4327623 DOI: 10.3389/fnana.2015.00010] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
The glial cell line-derived neurotrophic factor (GDNF) is a well-established trophic agent for dopaminergic (DA) neurons in vitro and in vivo. GDNF is necessary for maintenance of neuronal morphological and neurochemical phenotype and protects DA neurons from toxic damage. Numerous studies on animal models of Parkinson’s disease (PD) have reported beneficial effects of GDNF on nigrostriatal DA neuron survival. However, translation of these observations to the clinical setting has been hampered so far by side effects associated with the chronic continuous intra-striatal infusion of recombinant GDNF. In addition, double blind and placebo-controlled clinical trials have not reported any clinically relevant effect of GDNF on PD patients. In the past few years, experiments with conditional Gdnf knockout mice have suggested that GDNF is necessary for maintenance of DA neurons in adulthood. In parallel, new methodologies for exogenous GDNF delivery have been developed. Recently, it has been shown that a small population of scattered, electrically interconnected, parvalbumin positive (PV+) GABAergic interneurons is responsible for most of the GDNF produced in the rodent striatum. In addition, cholinergic striatal interneurons appear to be also involved in the modulation of striatal GDNF. In this review, we summarize current knowledge on brain GDNF delivery, homeostasis, and its effects on nigrostriatal DA neurons. Special attention is paid to the therapeutic potential of endogenous GDNF stimulation in PD.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain ; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla Seville, Spain ; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid, Spain
| |
Collapse
|
43
|
Alamri A, Ughratdar I, Samuel M, Ashkan K. Deep brain stimulation of the subthalamic nucleus in Parkinson's disease 2003–2013: Where are we another 10 years on? Br J Neurosurg 2015; 29:319-28. [DOI: 10.3109/02688697.2014.997669] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
44
|
Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, Sessa A, Colasante G, Bartolomeo R, Massimino L, Ferroni S, Settembre C, Benfenati F, Broccoli V. Direct conversion of fibroblasts into functional astrocytes by defined transcription factors. Stem Cell Reports 2014; 4:25-36. [PMID: 25556566 PMCID: PMC4297873 DOI: 10.1016/j.stemcr.2014.12.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 01/04/2023] Open
Abstract
Direct cell reprogramming enables direct conversion of fibroblasts into functional neurons and oligodendrocytes using a minimal set of cell-lineage-specific transcription factors. This approach is rapid and simple, generating the cell types of interest in one step. However, it remains unknown whether this technology can be applied to convert fibroblasts into astrocytes, the third neural lineage. Astrocytes play crucial roles in neuronal homeostasis, and their dysfunctions contribute to the origin and progression of multiple human diseases. Herein, we carried out a screening using several transcription factors involved in defining the astroglial cell fate and identified NFIA, NFIB, and SOX9 to be sufficient to convert with high efficiency embryonic and postnatal mouse fibroblasts into astrocytes (iAstrocytes). We proved both by gene-expression profiling and functional tests that iAstrocytes are comparable to native brain astrocytes. This protocol can be then employed to generate functional iAstrocytes for a wide range of experimental applications. NFIA, NFIB, and SOX9 reprogram fibroblasts into induced astrocytes (iAstrocytes) iAstrocytes reprogramming induces a global change in gene-expression profiling iAstrocytes are functionally comparable to native astrocytes NFIA, NFIB, and SOX9 induce an astrocytic phenotype in human fibroblasts
Collapse
Affiliation(s)
- Massimiliano Caiazzo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| | - Serena Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Pierluigi Valente
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy
| | - Gabriele Lignani
- Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | | | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rosa Bartolomeo
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, Naples 80131, Italy; Dulbecco Telethon Institute; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Medical Genetics, Department of Medical and Translational Science Unit, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Fabio Benfenati
- Section of Physiology, Department of Experimental Medicine, University of Genoa and National Institute of Neuroscience, 16132 Genoa, Italy; Department of Neuroscience and Brain Technologies, Italian Institute of Technology, 16132 Genoa, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan 20132, Italy.
| |
Collapse
|
45
|
Wang X, Zhao Z, Gong J, Zhou S, Peng H, Shatara A, Zhu TZ, Meltzer R, Du Y, Gu H. Adipose stem cells-conditioned medium blocks 6-hydroxydopamine-induced neurotoxicity via the IGF-1/PI3K/AKT pathway. Neurosci Lett 2014; 581:98-102. [PMID: 25161124 DOI: 10.1016/j.neulet.2014.08.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/31/2014] [Accepted: 08/18/2014] [Indexed: 12/22/2022]
Abstract
Previous studies suggest that the delivery of neurotrophic factors secreted from adipose stromal cells (ASC) protect the brain from 6-hydroxydopamine (6-OHDA)-induced neurotoxicity. However, it remains unclear which secreted neurotrophic factor has an important role in protecting 6-OHDA-treated neurons. Through the use of antibodies in this study, we demonstrated that specific neutralization of IGF-1 activity in ASC conditioned media (ASC-CM) significantly blocks ASC-CM-induced neuroprotection against 6-OHDA neurotoxicity. Consistently, this neuroprotection was mostly attributed to the activation of the AKT-mediated signaling pathway. In contrast, brain derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) in ASC-CM did not play a role in ASC-CM-induced neuroprotection against 6-OHDA.
Collapse
Affiliation(s)
- Xianjun Wang
- Department of Neurology, Linyi People's Hospital, No. 49 Yizhou Road, Linyi, Shandong 276000, P.R. China
| | - Zhenyu Zhao
- Department of Neurology, Linyi People's Hospital, No. 49 Yizhou Road, Linyi, Shandong 276000, P.R. China
| | - Jian Gong
- Department of Neurology, Linyi People's Hospital, No. 49 Yizhou Road, Linyi, Shandong 276000, P.R. China
| | - Shengnian Zhou
- Department of Neurology, Qilu Hospital of Shandong University and Bain Science Research Institute, Shandong University, No. 107 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| | - Hongjun Peng
- Department of Pediatrics, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, P.R. China.
| | - Adam Shatara
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Timmy Z Zhu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rebecca Meltzer
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
46
|
Cabezas R, Avila M, Gonzalez J, El-Bachá RS, Báez E, García-Segura LM, Jurado Coronel JC, Capani F, Cardona-Gomez GP, Barreto GE. Astrocytic modulation of blood brain barrier: perspectives on Parkinson's disease. Front Cell Neurosci 2014; 8:211. [PMID: 25136294 PMCID: PMC4120694 DOI: 10.3389/fncel.2014.00211] [Citation(s) in RCA: 295] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/14/2014] [Indexed: 12/21/2022] Open
Abstract
The blood–brain barrier (BBB) is a tightly regulated interface in the Central Nervous System (CNS) that regulates the exchange of molecules in and out from the brain thus maintaining the CNS homeostasis. It is mainly composed of endothelial cells (ECs), pericytes and astrocytes that create a neurovascular unit (NVU) with the adjacent neurons. Astrocytes are essential for the formation and maintenance of the BBB by providing secreted factors that lead to the adequate association between the cells of the BBB and the formation of strong tight junctions. Under neurological disorders, such as chronic cerebral ischemia, brain trauma, Epilepsy, Alzheimer and Parkinson’s Diseases, a disruption of the BBB takes place, involving a lost in the permeability of the barrier and phenotypical changes in both the ECs and astrocytes. In this aspect, it has been established that the process of reactive gliosis is a common feature of astrocytes during BBB disruption, which has a detrimental effect on the barrier function and a subsequent damage in neuronal survival. In this review we discuss the implications of astrocyte functions in the protection of the BBB, and in the development of Parkinson’s disease (PD) and related disorders. Additionally, we highlight the current and future strategies in astrocyte protection aimed at the development of restorative therapies for the BBB in pathological conditions.
Collapse
Affiliation(s)
- Ricardo Cabezas
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Marcos Avila
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Janneth Gonzalez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | | | - Eliana Báez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | | | - Juan Camilo Jurado Coronel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal, Facultad de Medicina, Instituto de Investigaciones cardiológicas Prof. Dr. Alberto C. Taquini (ININCA), UBA-CONICET, Buenos Aires Argentina
| | - Gloria Patricia Cardona-Gomez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Faculty of Medicine, SIU, University of Antioquia UdeA Medellín, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, D.C., Colombia
| |
Collapse
|