1
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
2
|
Zeini S, Davoodian N, Mousavi SA. Gamma-oryzanol attenuates lipopolysaccharide-induced cognitive impairment by modulation of hippocampal inflammatory response and glial activation in mice. J Neuroimmunol 2024; 387:578292. [PMID: 38278081 DOI: 10.1016/j.jneuroim.2024.578292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/26/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Systemic inflammation can cause chronic neuroinflammation, which is a significant risk factor for neurodegenerative disorders. Therefore, anti-inflammatory agents that reduce peripheral inflammation are potential targets for the prevention or treatment of these debilitating diseases. In the present study, we investigated whether gamma-oryzanol (ORY) could protect against chronic neuroinflammation induced by lipopolysaccharide (LPS) in adult male mice. Mice were injected with LPS (0.75 mg/kg/day) or saline for 7 consecutive days and orally received ORY (100 mg/kg) or vehicle for 14 days (7 days before LPS injections and 7 days co-treated with LPS). After two weeks, mice were subjected to behavioral assessments using the Morris water maze and Y-maze. Moreover, the expression level of several inflammatory mediators was measured in the hippocampus of treated animals. Also, neuronal loss, microglia, and astrocyte densities were evaluated in the CA1 and CA3 hippocampus. We found that ORY treatment significantly improved spatial and working memory in LPS-treated mice. This behavioral improvement was accompanied by a significant reduction in the number of microglia and astrocytes in the CA1 and CA3 hippocampus. Moreover, ORY treatment effectively prevented LPS-induced increases in the expression of inflammatory mediators and enhanced neuronal survival in the CA1 hippocampus. Our findings suggest that ORY treatment can be a therapeutic option to improve cognitive impairments and neuroinflammation induced by endotoxins.
Collapse
Affiliation(s)
- Shiva Zeini
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nahid Davoodian
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Seyed Abdollah Mousavi
- Pathology Department, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
3
|
Szlufik S, Kopeć K, Szleszkowski S, Koziorowski D. Glymphatic System Pathology and Neuroinflammation as Two Risk Factors of Neurodegeneration. Cells 2024; 13:286. [PMID: 38334678 PMCID: PMC10855155 DOI: 10.3390/cells13030286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/26/2024] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
The key to the effective treatment of neurodegenerative disorders is a thorough understanding of their pathomechanism. Neurodegeneration and neuroinflammation are mutually propelling brain processes. An impairment of glymphatic system function in neurodegeneration contributes to the progression of pathological processes. The question arises as to how neuroinflammation and the glymphatic system are related. This review highlights the direct and indirect influence of these two seemingly independent processes. Protein aggregates, a characteristic feature of neurodegeneration, are correlated with glymphatic clearance and neuroinflammation. Glial cells cannot be overlooked when considering the neuroinflammatory processes. Astrocytes are essential for the effective functioning of the glymphatic system and play a crucial role in the inflammatory responses in the central nervous system. It is imperative to acknowledge the significance of AQP4, a protein that exhibits a high degree of polarization in astrocytes and is crucial for the functioning of the glymphatic system. AQP4 influences inflammatory processes that have not yet been clearly delineated. Another interesting issue is the gut-brain axis and microbiome, which potentially impact the discussed processes. A discussion of the correlation between the functioning of the glymphatic system and neuroinflammation may contribute to exploring the pathomechanism of neurodegeneration.
Collapse
Affiliation(s)
- Stanisław Szlufik
- Department of Neurology, Faculty of Health Science, Medical University of Warsaw, 02-091 Warszawa, Poland; (K.K.)
| | | | | | | |
Collapse
|
4
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
5
|
Ramakrishnan P, Joshi A, Fazil M, Yadav P. A comprehensive review on therapeutic potentials of photobiomodulation for neurodegenerative disorders. Life Sci 2024; 336:122334. [PMID: 38061535 DOI: 10.1016/j.lfs.2023.122334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
A series of experimental trials over the past two centuries has put forth Photobiomodulation (PBM) as a treatment modality that utilizes colored lights for various conditions. While in its cradle, PBM was used for treating simple conditions such as burns and wounds, advancements in recent years have extended the use of PBM for treating complex neurodegenerative diseases (NDDs). PBM has exhibited the potential to curb several symptoms and signs associated with NDDs. While several of the currently used therapeutics cause adverse side effects alongside being highly invasive, PBM on the contrary, seems to be broad-acting, less toxic, and non-invasive. Despite being projected as an ideal therapeutic for NDDs, PBM still isn't considered a mainstream treatment modality due to some of the challenges and knowledge gaps associated with it. Here, we review the advantages of PBM summarized above with an emphasis on the common mechanisms that underlie major NDDs and how PBM helps tackle them. We also discuss important questions such as whether PBM should be considered a mainstay treatment modality for these conditions and if PBM's properties can be harnessed to develop prophylactic therapies for high-risk individuals and also highlight important animal studies that underscore the importance of PBM and the challenges associated with it. Overall, this review is intended to bring the major advances made in the field to the spotlight alongside addressing the practicalities and caveats to develop PBM as a major therapeutic for NDDs.
Collapse
Affiliation(s)
- Pooja Ramakrishnan
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| | - Aradhana Joshi
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| | - Mohamed Fazil
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India; School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India
| | - Pankaj Yadav
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| |
Collapse
|
6
|
Ma YM, Zhao L. Mechanism and Therapeutic Prospect of miRNAs in Neurodegenerative Diseases. Behav Neurol 2023; 2023:8537296. [PMID: 38058356 PMCID: PMC10697780 DOI: 10.1155/2023/8537296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 08/30/2023] [Accepted: 10/21/2023] [Indexed: 12/08/2023] Open
Abstract
MicroRNAs (miRNAs) are the smallest class of noncoding RNAs, which widely exist in animals and plants. They can inhibit translation or overexpression by combining with mRNA and participate in posttranscriptional regulation of genes, resulting in reduced expression of target proteins, affecting the development, growth, aging, metabolism, and other physiological and pathological processes of animals and plants. It is a powerful negative regulator of gene expression. It mediates the information exchange between different cellular pathways in cellular homeostasis and stress response and regulates the differentiation, plasticity, and neurotransmission of neurons. In neurodegenerative diseases, in addition to the complex interactions between genetic susceptibility and environmental factors, miRNAs can serve as a promising diagnostic tool for diseases. They can also increase or reduce neuronal damage by regulating the body's signaling pathways, immune system, stem cells, gut microbiota, etc. They can not only affect the occurrence of diseases and exacerbate disease progression but also promote neuronal repair and reduce apoptosis, to prevent and slow down the development of diseases. This article reviews the research progress of miRNAs on the mechanism and treatment of neurodegenerative diseases in the nervous system. This trial is registered with NCT01819545, NCT02129452, NCT04120493, NCT04840823, NCT02253732, NCT02045056, NCT03388242, NCT01992029, NCT04961450, NCT03088839, NCT04137926, NCT02283073, NCT04509271, NCT02859428, and NCT05243017.
Collapse
Affiliation(s)
- Ya-Min Ma
- Acupuncture and Massage Department of Nanyang Traditional Chinese Medicine Hospital, Wo Long District, Nanyang City 473000, China
| | - Lan Zhao
- Tianjin Key Laboratory of Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Xiqing District, Tianjin 300381, China
| |
Collapse
|
7
|
Kaikai NE, Ba-M'hamed S, Ghanima A, Bennis M. Exposure to metam sodium-based pesticide impaired cognitive performances in adult mice: Involvement of oxidative damage and glial activation. Toxicol Appl Pharmacol 2023; 477:116677. [PMID: 37678439 DOI: 10.1016/j.taap.2023.116677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/28/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Cognitive integrity is a critical aspect of neurological function, and a decline in cognitive function is a hallmark of neurotoxicity. Oxidative stress is a significant pathological feature contributing to cognitive deficits that can arise from exposure to environmental pollutants such as pesticides. Among these, Metam sodium-based pesticides (MS-BP) are an emergent type of pesticide widely used in the agriculture and public health sectors for controlling pests and diseases. Our prior research has shown that animals exposed to MS-BP during the early stages of brain development caused cognitive impairments. In the present study, we tested whether exposure to this compound in a fully matured brain would affect cognitive performance and induce oxidative damage to the central nervous system. In this context, adult mice received chronic treatment with increasing doses of MS-BP and subjected to a set of behavioral paradigms. Following behavioral assessment, oxidative stress and glial activation were evaluated. Our main findings showed that MS-BP chronic exposure impaired recognition and short- and long-term memory. These alterations were accompanied by increased superoxide dismutase activity and malondialdehyde level and a marked decrease in catalase activity in specific brain areas. Moreover, exposure to MS-BP is associated with a significant rise in the density of astrocytic and microglial markers, indicating a possible glial cell response within the prefrontal cortex and hippocampus. The present work demonstrated that MS-BP altered cognitive performance likely through oxidative damage to the brain.
Collapse
Affiliation(s)
- Nour-Eddine Kaikai
- Laboratory of Pharmacology, Neurobiology, Anthropology, and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco; Department of Biology, Higher Normal School, Cadi Ayyad University, 4000 Marrakech, Morocco
| | - Saadia Ba-M'hamed
- Laboratory of Pharmacology, Neurobiology, Anthropology, and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco
| | - Abderrazzak Ghanima
- Research Laboratory for Sustainable Development and Health, Cadi Ayyad University, Faculty of Sciences and Techniques, Marrakech, Morocco
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropology, and Environment, Cadi Ayyad University, Faculty of Sciences, Marrakech, Morocco.
| |
Collapse
|
8
|
Stöberl N, Maguire E, Salis E, Shaw B, Hall-Roberts H. Human iPSC-derived glia models for the study of neuroinflammation. J Neuroinflammation 2023; 20:231. [PMID: 37817184 PMCID: PMC10566197 DOI: 10.1186/s12974-023-02919-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023] Open
Abstract
Neuroinflammation is a complex biological process that plays a significant role in various brain disorders. Microglia and astrocytes are the key cell types involved in inflammatory responses in the central nervous system. Neuroinflammation results in increased levels of secreted inflammatory factors, such as cytokines, chemokines, and reactive oxygen species. To model neuroinflammation in vitro, various human induced pluripotent stem cell (iPSC)-based models have been utilized, including monocultures, transfer of conditioned media between cell types, co-culturing multiple cell types, neural organoids, and xenotransplantation of cells into the mouse brain. To induce neuroinflammatory responses in vitro, several stimuli have been established that can induce responses in either microglia, astrocytes, or both. Here, we describe and critically evaluate the different types of iPSC models that can be used to study neuroinflammation and highlight how neuroinflammation has been induced and measured in these cultures.
Collapse
Affiliation(s)
- Nina Stöberl
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Emily Maguire
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Elisa Salis
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Bethany Shaw
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| | - Hazel Hall-Roberts
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff, CF10 3AT UK
| |
Collapse
|
9
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
10
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Valori CF, Sulmona C, Brambilla L, Rossi D. Astrocytes: Dissecting Their Diverse Roles in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells 2023; 12:1450. [PMID: 37296571 PMCID: PMC10252425 DOI: 10.3390/cells12111450] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are fatal neurodegenerative disorders often co-occurring in the same patient, a feature that suggests a common origin of the two diseases. Consistently, pathological inclusions of the same proteins as well as mutations in the same genes can be identified in both ALS/FTD. Although many studies have described several disrupted pathways within neurons, glial cells are also regarded as crucial pathogenetic contributors in ALS/FTD. Here, we focus our attention on astrocytes, a heterogenous population of glial cells that perform several functions for optimal central nervous system homeostasis. Firstly, we discuss how post-mortem material from ALS/FTD patients supports astrocyte dysfunction around three pillars: neuroinflammation, abnormal protein aggregation, and atrophy/degeneration. Furthermore, we summarize current attempts at monitoring astrocyte functions in living patients using either novel imaging strategies or soluble biomarkers. We then address how astrocyte pathology is recapitulated in animal and cellular models of ALS/FTD and how we used these models both to understand the molecular mechanisms driving glial dysfunction and as platforms for pre-clinical testing of therapeutics. Finally, we present the current clinical trials for ALS/FTD, restricting our discussion to treatments that modulate astrocyte functions, directly or indirectly.
Collapse
Affiliation(s)
- Chiara F. Valori
- Molecular Neuropathology of Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), 72072 Tübingen, Germany
- Department of Neuropathology, University of Tübingen, 72076 Tübingen, Germany
| | - Claudia Sulmona
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| |
Collapse
|
12
|
Ahmed M, Muffat J, Li Y. Understanding neural development and diseases using CRISPR screens in human pluripotent stem cell-derived cultures. Front Cell Dev Biol 2023; 11:1158373. [PMID: 37101616 PMCID: PMC10123288 DOI: 10.3389/fcell.2023.1158373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023] Open
Abstract
The brain is arguably the most complex part of the human body in form and function. Much remains unclear about the molecular mechanisms that regulate its normal and pathological physiology. This lack of knowledge largely stems from the inaccessible nature of the human brain, and the limitation of animal models. As a result, brain disorders are difficult to understand and even more difficult to treat. Recent advances in generating human pluripotent stem cells (hPSCs)-derived 2-dimensional (2D) and 3-dimensional (3D) neural cultures have provided an accessible system to model the human brain. Breakthroughs in gene editing technologies such as CRISPR/Cas9 further elevate the hPSCs into a genetically tractable experimental system. Powerful genetic screens, previously reserved for model organisms and transformed cell lines, can now be performed in human neural cells. Combined with the rapidly expanding single-cell genomics toolkit, these technological advances culminate to create an unprecedented opportunity to study the human brain using functional genomics. This review will summarize the current progress of applying CRISPR-based genetic screens in hPSCs-derived 2D neural cultures and 3D brain organoids. We will also evaluate the key technologies involved and discuss their related experimental considerations and future applications.
Collapse
Affiliation(s)
- Mai Ahmed
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Cheng C, Wan H, Cong P, Huang X, Wu T, He M, Zhang Q, Xiong L, Tian L. Targeting neuroinflammation as a preventive and therapeutic approach for perioperative neurocognitive disorders. J Neuroinflammation 2022; 19:297. [PMID: 36503642 PMCID: PMC9743533 DOI: 10.1186/s12974-022-02656-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Perioperative neurocognitive disorders (PND) is a common postoperative complication associated with regional or general anesthesia and surgery. Growing evidence in both patient and animal models of PND suggested that neuroinflammation plays a critical role in the development and progression of this problem, therefore, mounting efforts have been made to develop novel therapeutic approaches for PND by targeting specific factors or steps alongside the neuroinflammation. Multiple studies have shown that perioperative anti-neuroinflammatory strategies via administering pharmacologic agents or performing nonpharmacologic approaches exert benefits in the prevention and management of PND, although more clinical evidence is urgently needed to testify or confirm these results. Furthermore, long-term effects and outcomes with respect to cognitive functions and side effects are needed to be observed. In this review, we discuss recent preclinical and clinical studies published within a decade as potential preventive and therapeutic approaches targeting neuroinflammation for PND.
Collapse
Affiliation(s)
- Chun Cheng
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Hanxi Wan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Peilin Cong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Xinwei Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Tingmei Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Mengfan He
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Qian Zhang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| | - Li Tian
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, 200434 China ,grid.24516.340000000123704535Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, 200434 China ,grid.24516.340000000123704535Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, 200434 China
| |
Collapse
|
14
|
Della Vecchia S, Marchese M, Santorelli FM. Glial Contributions to Lafora Disease: A Systematic Review. Biomedicines 2022; 10:biomedicines10123103. [PMID: 36551859 PMCID: PMC9776290 DOI: 10.3390/biomedicines10123103] [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: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Lafora disease (LD) is a neurodegenerative condition characterized by the accumulation of polyglucosan bodies (PBs) throughout the brain. Alongside metabolic and molecular alterations, neuroinflammation has emerged as another key histopathological feature of LD. METHODS To investigate the role of astrocytes and microglia in LD, we performed a systematic review according to the PRISMA statement. PubMed, Scopus, and Web-of-Science database searches were performed independently by two reviewers. RESULTS Thirty-five studies analyzing the relationship of astrocytes and microglia with LD and/or the effects of anti-inflammatory treatments in LD animal models were identified and included in the review. Although LD has long been dominated by a neuronocentric view, a growing body of evidence suggests a role of glial cells in the disease, starting with the finding that these cells accumulate PBs. We discuss the potential meaning of glial PB accumulations, the likely factors activating glial cells, and the possible contribution of glial cells to LD neurodegeneration and epilepsy. CONCLUSIONS Given the evidence for the role of neuroinflammation in LD, future studies should consider glial cells as a potential therapeutic target for modifying/delaying LD progression; however, it should be kept in mind that these cells can potentially assume multiple reactive phenotypes, which could influence the therapeutic response.
Collapse
Affiliation(s)
- Stefania Della Vecchia
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Correspondence: (S.D.V.); (F.M.S.)
| | - Maria Marchese
- Neurobiology, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Neurobiology, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Correspondence: (S.D.V.); (F.M.S.)
| |
Collapse
|
15
|
Boucher T, Liang S, Brown AM. Advancing basic and translational research to deepen understanding of the molecular immune-mediated mechanisms regulating long-term persistence of HIV-1 in microglia in the adult human brain. J Leukoc Biol 2022; 112:1223-1231. [PMID: 35612272 PMCID: PMC9613482 DOI: 10.1002/jlb.1mr0422-620r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/22/2022] [Indexed: 12/30/2022] Open
Abstract
Knowledge about the diversity microglia (MG) type and function in the rodent and human brain has advanced significantly in the last few years. Nevertheless, we have known for 40 years that MG, monocytes, and macrophages in the brain play crucial roles in the pathogenesis of the HIV-1 in all tissues. HIV enters and spreads in the brain early, long before the initiation of antiviral therapy. As a result, many people with HIV continue to experience neurologic and neuropsychiatric comorbid conditions collectively known as HIV-associated neurocognitive disorder (HAND). HIV pathogenic sequelae in the CNS pose a challenge for cure strategies. Detailed understanding at a mechanistic level of how low-level and latent HIV-1 infection in MG negatively impacts neuroglial function has remained somewhat elusive. Direct rigorous in vivo experimental validation that the virus can integrate into MG and assume a latent but reactivatable state has remained constrained. However, there is much excitement that human in vitro models for MG can now help close the gap. This review will provide a brief background to place the role of MG in the ongoing neurologic complications of HIV infection of the CNS, then focus on the use and refinement of human postmitotic monocyte-derived MG-like cells and how they are being applied to advance research on HIV persistence and proinflammatory signaling in the CNS. Critically, an understanding of myeloid plasticity and heterogeneity and rigorous attention to all aspects of cell handling is essential for reproducibility. Summary Sentence: This review focuses on human postmitotic monocyte-derived microglia-like cells as tools to advance research on HIV persistence and neuroinflammatory signaling.
Collapse
Affiliation(s)
- Thomas Boucher
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Shijun Liang
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Amanda M. Brown
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
16
|
Poulot-Becq-Giraudon Y, Carrillo-de Sauvage MA, Escartin C. Astrocytes réactifs et maladies cérébrales. Med Sci (Paris) 2022; 38:786-794. [DOI: 10.1051/medsci/2022104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Les astrocytes sont des partenaires essentiels des neurones dans le système nerveux central. En réponse à de nombreuses maladies qui touchent le cerveau, les astrocytes subissent des modifications morphologiques, moléculaires et fonctionnelles : ils deviennent réactifs. Ces changements multiples sont susceptibles d’avoir un impact important sur les neurones, qui dépendent de nombreuses fonctions remplies par les astrocytes. La réponse de réactivité astrocytaire dépend du contexte pathologique. Il est donc indispensable de définir précisément les changements qui se produisent dans les astrocytes réactifs dans chaque situation pathologique, par des approches adaptées et sélectives. Cela permettra le développement de thérapies innovantes ciblant ces cellules partenaires des neurones, ainsi que l’identification de biomarqueurs spécifiques de certaines maladies cérébrales.
Collapse
|
17
|
Ongnok B, Maneechote C, Chunchai T, Pantiya P, Arunsak B, Nawara W, Chattipakorn N, Chattipakorn SC. Modulation of mitochondrial dynamics rescues cognitive function in rats with 'doxorubicin-induced chemobrain' via mitigation of mitochondrial dysfunction and neuroinflammation. FEBS J 2022; 289:6435-6455. [PMID: 35514149 DOI: 10.1111/febs.16474] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/26/2022] [Accepted: 05/04/2022] [Indexed: 01/18/2023]
Abstract
Doxorubicin (DOX), an effective, extensively used chemotherapeutic drug, can cause cognitive deterioration in cancer patients. The associated debilitating neurological sequelae are referred to as chemobrain. Our recent work demonstrated that Dox treatment resulted in an imbalance in mitochondrial dynamics, ultimately culminating in cognitive decline in rats. Therefore, in this study, we aim to explore the therapeutic efficacy of a pharmacological intervention, which modulates mitochondrial dynamics using a potent mitochondrial fission inhibitor (Mdivi-1) and mitochondrial fusion promoter (M1) against Dox-induced chemobrain. In the study, male Wistar rats were randomly assigned to receive either normal saline solution or six doses of Dox (3 mg·kg-1 ) via intraperitoneal injection. Then, the Dox-treated rats were intraperitoneally given either 1% DMSO as the vehicle, Mdivi-1 (1.2 mg·kg-1 ), M1 (2 mg·kg-1 ), or a combined treatment of Mdivi-1 and M1 for 30 consecutive days. Long-term learning and memory were evaluated using the novel object location task and novel object recognition task. Following euthanasia, the rat brains were dissected to enable further molecular investigation. We demonstrated that long-term treatment with mitochondrial dynamic modulators suppressed mitochondrial fission in the hippocampus following Dox treatment, leading to an improvement in brain homeostasis. Mitochondrial dynamic modulator treatments restored cognitive function in Dox-treated rats by attenuating neuroinflammation, decreasing oxidative stress, preserving synaptic integrity, reducing potential Alzheimer's related lesions, and mitigating both apoptosis and necroptosis following Dox administration. Together, our findings suggested that mitochondrial dynamics modulators protected against Dox-induced cognitive impairment by rebalancing mitochondrial homeostasis and attenuating both oxidative and inflammatory insults.
Collapse
Affiliation(s)
- Benjamin Ongnok
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Thailand
| | - Chayodom Maneechote
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
| | - Titikorn Chunchai
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
| | - Patcharapong Pantiya
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Thailand
| | - Busarin Arunsak
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
| | - Wichwara Nawara
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
| | - Nipon Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Thailand
| | - Siriporn C Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Thailand
| |
Collapse
|
18
|
Liu D, Ji Q, Cheng Y, Liu M, Zhang B, Mei Q, Huan M, Zhou S. Cyclosporine A loaded brain targeting nanoparticle to treat cerebral ischemia/reperfusion injury in mice. J Nanobiotechnology 2022; 20:256. [PMID: 35658867 PMCID: PMC9164331 DOI: 10.1186/s12951-022-01474-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/23/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Ischemic stroke is one of the main causes of death and disability in the world. The treatment for ischemic stroke is to restore blood perfusion as soon as possible. However, when ischemic brain tissue is re-perfused by blood, the mitochondrial permeability transition pore (mPTP) in neuron and microglia is excessively opened, resulting in the apoptosis of neuron and nerve inflammation. This aggravates nerve injury. Cyclosporine A (CsA) inhibits the over-opening of mPTP, subsequently reducing the release of ROS and the apoptosis of cerebral ischemia/reperfusion injured neuron and microglia. However, CsA is insoluble in water and present in high concentrations in lymphatic tissue. Herein, cerebral infarction tissue targeted nanoparticle (CsA@HFn) was developed to treat cerebral ischemia/reperfusion injury. RESULTS CsA@HFn efficiently penetrated the blood-brain barrier (BBB) and selectively accumulated in ischemic area, inhibiting the opening of mPTP and ROS production in neuron. This subsequently reduced the apoptosis of neuron and the damage of BBB. Consequently, CsA@HFn significantly reduced the infarct area. Moreover, CsA@HFn inhibited the recruitment of astrocytes and microglia in ischemic region and polarized microglia into M2 type microglia, which subsequently alleviated the nerve inflammation. CONCLUSIONS CsA@HFn showed a significant therapeutic effect on cerebral ischemia/reperfusion injury by alleviating the apoptosis of neuron, nerve inflammation and the damage of BBB in ischemic area. CsA@HFn has great potential in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Daozhou Liu
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Qifeng Ji
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Ying Cheng
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Miao Liu
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Bangle Zhang
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Qibing Mei
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Menglei Huan
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Siyuan Zhou
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| |
Collapse
|
19
|
Indari O, Jakhmola S, Pathak DK, Tanwar M, Kandpal M, Mishra A, Kumar R, Jha HC. Comparative Account of Biomolecular Changes Post Epstein Barr Virus Infection of the Neuronal and Glial Cells Using Raman Microspectroscopy. ACS Chem Neurosci 2022; 13:1627-1637. [PMID: 35561419 DOI: 10.1021/acschemneuro.2c00081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Raman microspectroscopy is a vibrational spectroscopy technique used for investigating molecular fingerprints of a wide range of liquid or solid samples. The technique can be efficiently utilized to understand the virus-mediated cellular changes and could provide valuable insights into specific biomolecular alterations. The Epstein Barr virus (EBV) has been associated with various types of cancers as well as neurodegenerative diseases. However, EBV-mediated neurological ailments are yet underexplored in terms of biomolecular changes in neuronal and glial cells (astrocytes and microglia). In continuation of our earlier exploration of EBV-influenced glial cells, we tried to decipher biomolecular changes in EBV-infected neuronal cells using Raman microspectroscopy. Additionally, we compared the consecutive biomolecular changes observed in neuronal cells with both the glial cells. We observed that EBV infection gets differentially regulated in the neuronal cells, astrocytes, and microglia. The viral entry and initiation of infection-mediated cellular modulation could start as soon as 2 h post infection but may regulate a distinct biomolecular milieu in different time intervals. Similar to the early timespan, the 24-36 h interval could also be important for EBV to manipulate neuronal as well as glial cells as depicted from elevated biomolecular activities. At these time intervals, some common biomolecules such as proline, glucose, lactic acid, nucleotides, or cholesterol were observed in the cells. However, at these time intervals, some distinct biomolecules were also observed in each cell, such as collagen, lipid, and protein stretches in the neuronal nucleus (2-4 h); tyrosine and RNA in the astrocyte nucleus (2-4 h nucleus); and fatty acids in the microglia nucleus (24-36 h). The observed biomolecular entities could ultimately play pivotal roles in the viral usurpation of cells. We also provided insights into whether these biomolecular changes can be correlated to each other and mediate virus-associated manifestations which can be linked to neurological complications. Our study aids in the understanding of EBV-mediated biomolecular changes in the various compartments of the central nervous system.
Collapse
Affiliation(s)
- Omkar Indari
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Shweta Jakhmola
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Devesh K. Pathak
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
- Department of Chemical Engineering, University of Seoul, Seoul 02504, Republic of Korea
| | - Manushree Tanwar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur 342011, India
| | - Rajesh Kumar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| |
Collapse
|
20
|
Labib D, Wang Z, Prakash P, Zimmer M, Smith MD, Frazel PW, Barbar L, Sapar ML, Calabresi PA, Peng J, Liddelow SA, Fossati V. Proteomic Alterations and Novel Markers of Neurotoxic Reactive Astrocytes in Human Induced Pluripotent Stem Cell Models. Front Mol Neurosci 2022; 15:870085. [PMID: 35592112 PMCID: PMC9113221 DOI: 10.3389/fnmol.2022.870085] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/29/2022] [Indexed: 12/20/2022] Open
Abstract
Astrocytes respond to injury, infection, and inflammation in the central nervous system by acquiring reactive states in which they may become dysfunctional and contribute to disease pathology. A sub-state of reactive astrocytes induced by proinflammatory factors TNF, IL-1α, and C1q ("TIC") has been implicated in many neurodegenerative diseases as a source of neurotoxicity. Here, we used an established human induced pluripotent stem cell (hiPSC) model to investigate the surface marker profile and proteome of TIC-induced reactive astrocytes. We propose VCAM1, BST2, ICOSL, HLA-E, PD-L1, and PDPN as putative, novel markers of this reactive sub-state. We found that several of these markers colocalize with GFAP+ cells in post-mortem samples from people with Alzheimer's disease. Moreover, our whole-cells proteomic analysis of TIC-induced reactive astrocytes identified proteins and related pathways primarily linked to potential engagement with peripheral immune cells. Taken together, our findings will serve as new tools to purify reactive astrocyte subtypes and to further explore their involvement in immune responses associated with injury and disease.
Collapse
Affiliation(s)
- David Labib
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Priya Prakash
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
| | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Paul W. Frazel
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Maria L. Sapar
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, United States
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, United States
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, United States
| |
Collapse
|
21
|
Harry GJ, McBride S, Witchey SK, Mhaouty-Kodja S, Trembleau A, Bridge M, Bencsik A. Roadbumps at the Crossroads of Integrating Behavioral and In Vitro Approaches for Neurotoxicity Assessment. FRONTIERS IN TOXICOLOGY 2022; 4:812863. [PMID: 35295216 PMCID: PMC8915899 DOI: 10.3389/ftox.2022.812863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/25/2022] [Indexed: 12/15/2022] Open
Abstract
With the appreciation that behavior represents the integration and complexity of the nervous system, neurobehavioral phenotyping and assessment has seen a renaissance over the last couple of decades, resulting in a robust database on rodent performance within various testing paradigms, possible associations with human disorders, and therapeutic interventions. The interchange of data across behavior and other test modalities and multiple model systems has advanced our understanding of fundamental biology and mechanisms associated with normal functions and alterations in the nervous system. While there is a demonstrated value and power of neurobehavioral assessments for examining alterations due to genetic manipulations, maternal factors, early development environment, the applied use of behavior to assess environmental neurotoxicity continues to come under question as to whether behavior represents a sensitive endpoint for assessment. Why is rodent behavior a sensitive tool to the neuroscientist and yet, not when used in pre-clinical or chemical neurotoxicity studies? Applying new paradigms and evidence on the biological basis of behavior to neurobehavioral testing requires expertise and refinement of how such experiments are conducted to minimize variability and maximize information. This review presents relevant issues of methods used to conduct such test, sources of variability, experimental design, data analysis, interpretation, and reporting. It presents beneficial and critical limitations as they translate to the in vivo environment and considers the need to integrate across disciplines for the best value. It proposes that a refinement of behavioral assessments and understanding of subtle pronounced differences will facilitate the integration of data obtained across multiple approaches and to address issues of translation.
Collapse
Affiliation(s)
- G. Jean Harry
- Neurotoxicology Group, Molecular Toxicology Branch, Division National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Sandra McBride
- Social & Scientific Systems, Inc., a DLH Holdings Company, Durham, NC, United States
| | - Shannah K. Witchey
- Division National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Paris, France
| | - Alain Trembleau
- Sorbonne Université, CNRS UMR8246, Inserm U1130, Institut de Biologie Paris Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Matthew Bridge
- Social & Scientific Systems, Inc., a DLH Holdings Company, Durham, NC, United States
| | - Anna Bencsik
- Anses Laboratoire de Lyon, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université de Lyon 1, Lyon, France
| |
Collapse
|
22
|
Allison RL, Welby E, Khayrullina G, Burnett BG, Ebert AD. Viral mediated knockdown of GATA6 in SMA iPSC-derived astrocytes prevents motor neuron loss and microglial activation. Glia 2022; 70:989-1004. [PMID: 35088910 PMCID: PMC9303278 DOI: 10.1002/glia.24153] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
Abstract
Spinal muscular atrophy (SMA), a pediatric genetic disorder, is characterized by the profound loss of spinal cord motor neurons and subsequent muscle atrophy and death. Although the mechanisms underlying motor neuron loss are not entirely clear, data from our work and others support the idea that glial cells contribute to disease pathology. GATA6, a transcription factor that we have previously shown to be upregulated in SMA astrocytes, is negatively regulated by SMN (survival motor neuron) and can increase the expression of inflammatory regulator NFκB. In this study, we identified upregulated GATA6 as a contributor to increased activation, pro-inflammatory ligand production, and neurotoxicity in spinal-cord patterned astrocytes differentiated from SMA patient induced pluripotent stem cells. Reducing GATA6 expression in SMA astrocytes via lentiviral infection ameliorated these effects to healthy control levels. Additionally, we found that SMA astrocytes contribute to SMA microglial phagocytosis, which was again decreased by lentiviral-mediated knockdown of GATA6. Together these data identify a role of GATA6 in SMA astrocyte pathology and further highlight glia as important targets of therapeutic intervention in SMA.
Collapse
Affiliation(s)
- Reilly L Allison
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Emily Welby
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Guzal Khayrullina
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland, USA
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, Maryland, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
23
|
Stifani S. Taking Cellular Heterogeneity Into Consideration When Modeling Astrocyte Involvement in Amyotrophic Lateral Sclerosis Using Human Induced Pluripotent Stem Cells. Front Cell Neurosci 2021; 15:707861. [PMID: 34602979 PMCID: PMC8485040 DOI: 10.3389/fncel.2021.707861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/24/2021] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are a large group of glial cells that perform a variety of physiological functions in the nervous system. They provide trophic, as well as structural, support to neuronal cells. Astrocytes are also involved in neuroinflammatory processes contributing to neuronal dysfunction and death. Growing evidence suggests important roles for astrocytes in non-cell autonomous mechanisms of motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Understanding these mechanisms necessitates the combined use of animal and human cell-based experimental model systems, at least in part because human astrocytes display a number of unique features that cannot be recapitulated in animal models. Human induced pluripotent stem cell (hiPSC)-based approaches provide the opportunity to generate disease-relevant human astrocytes to investigate the roles of these cells in ALS. These approaches are facing the growing recognition that there are heterogenous populations of astrocytes in the nervous system which are not functionally equivalent. This review will discuss the importance of taking astrocyte heterogeneity into consideration when designing hiPSC-based strategies aimed at generating the most informative preparations to study the contribution of astrocytes to ALS pathophysiology.
Collapse
Affiliation(s)
- Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
24
|
Reiss D, Maurin H, Audouard E, Martínez-Navarro M, Xue Y, Herault Y, Maldonado R, Cabañero D, Gaveriaux-Ruff C. Delta Opioid Receptor in Astrocytes Contributes to Neuropathic Cold Pain and Analgesic Tolerance in Female Mice. Front Cell Neurosci 2021; 15:745178. [PMID: 34602984 PMCID: PMC8483180 DOI: 10.3389/fncel.2021.745178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023] Open
Abstract
Background: The delta opioid receptor (DOR) contributes to pain control, and a major challenge is the identification of DOR populations that control pain, analgesia, and tolerance. Astrocytes are known as important cells in the pathophysiology of chronic pain, and many studies report an increased prevalence of pain in women. However, the implication of astrocytic DOR in neuropathic pain and analgesia, as well as the influence of sex in this receptor activity, remains unknown. Experimental Approach: We developed a novel conditional knockout (cKO) mouse line wherein DOR is deleted in astrocytes (named GFAP-DOR-KO), and investigated neuropathic mechanical allodynia as well as analgesia and analgesic tolerance in mutant male and female mice. Neuropathic cold allodynia was also characterized in mice of both sexes lacking DOR either in astrocytes or constitutively. Results: Neuropathic mechanical allodynia was similar in GFAP-DOR-KO and floxed DOR control mice, and the DOR agonist SNC80 produced analgesia in mutant mice of both sexes. Interestingly, analgesic tolerance developed in cKO males and was abolished in cKO females. Cold neuropathic allodynia was reduced in mice with decreased DOR in astrocytes. By contrast, cold allodynia was exacerbated in full DOR KO females. Conclusions: These findings show that astrocytic DOR has a prominent role in promoting cold allodynia and analgesic tolerance in females, while overall DOR activity was protective. Altogether this suggests that endogenous- and exogenous-mediated DOR activity in astrocytes worsens neuropathic allodynia while DOR activity in other cells attenuates this form of pain. In conclusion, our results show a sex-specific implication of astrocytic DOR in neuropathic pain and analgesic tolerance. These findings open new avenues for developing tailored DOR-mediated analgesic strategies.
Collapse
Affiliation(s)
- David Reiss
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Hervé Maurin
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Emilie Audouard
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Miriam Martínez-Navarro
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Yaping Xue
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández Elche, Alicante, Spain
| | - Claire Gaveriaux-Ruff
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
25
|
Kalkowski L, Golubczyk D, Kwiatkowska J, Holak P, Milewska K, Janowski M, Oliveira JM, Walczak P, Malysz-Cymborska I. Two in One: Use of Divalent Manganese Ions as Both Cross-Linking and MRI Contrast Agent for Intrathecal Injection of Hydrogel-Embedded Stem Cells. Pharmaceutics 2021; 13:pharmaceutics13071076. [PMID: 34371767 PMCID: PMC8309201 DOI: 10.3390/pharmaceutics13071076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/04/2022] Open
Abstract
Cell therapy is a promising tool for treating central nervous system (CNS) disorders; though, the translational efforts are plagued by ineffective delivery methods. Due to the large contact surface with CNS and relatively easy access, the intrathecal route of administration is attractive in extensive or global diseases such as stroke or amyotrophic lateral sclerosis (ALS). However, the precision and efficacy of this approach are still a challenge. Hydrogels were introduced to minimize cell sedimentation and improve cell viability. At the same time, contrast agents were integrated to allow image-guided injection. Here, we report using manganese ions (Mn2+) as a dual agent for cross-linking alginate-based hydrogels and magnetic resonance imaging (MRI). We performed in vitro studies to test the Mn2+ alginate hydrogel formulations for biocompatibility, injectability, MRI signal retention time, and effect on cell viability. The selected formulation was injected intrathecally into pigs under MRI control. The biocompatibility test showed a lack of immune response, and cells suspended in the hydrogel showed greater viability than monolayer culture. Moreover, Mn2+-labeled hydrogel produced a strong T1 MRI signal, which enabled MRI-guided procedure. We confirmed the utility of Mn2+ alginate hydrogel as a carrier for cells in large animals and a contrast agent at the same time.
Collapse
Affiliation(s)
- Lukasz Kalkowski
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Joanna Kwiatkowska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Piotr Holak
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Kamila Milewska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
| | - Miroslaw Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (M.J.); (P.W.)
| | - Joaquim Miguel Oliveira
- a3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| | - Piotr Walczak
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (M.J.); (P.W.)
| | - Izabela Malysz-Cymborska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (L.K.); (D.G.); (J.K.); (K.M.)
- Correspondence: ; Tel.: +48-605118887
| |
Collapse
|
26
|
Ongnok B, Khuanjing T, Chunchai T, Pantiya P, Kerdphoo S, Arunsak B, Nawara W, Jaiwongkam T, Apaijai N, Chattipakorn N, Chattipakorn SC. Donepezil Protects Against Doxorubicin-Induced Chemobrain in Rats via Attenuation of Inflammation and Oxidative Stress Without Interfering With Doxorubicin Efficacy. Neurotherapeutics 2021; 18:2107-2125. [PMID: 34312765 PMCID: PMC8608968 DOI: 10.1007/s13311-021-01092-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2021] [Indexed: 02/08/2023] Open
Abstract
Although doxorubicin (Dox) is an effective chemotherapy medication used extensively in the treatment of breast cancer, it frequently causes debilitating neurological deficits known as chemobrain. Donepezil (DPZ), an acetylcholinesterase inhibitor, provides therapeutic benefits in various neuropathological conditions. However, comprehensive mechanistic insights regarding the neuroprotection of DPZ on cognition and brain pathologies in a Dox-induced chemobrain model remain obscure. Here, we demonstrated that Dox-treated rats manifested conspicuous cognitive deficits and developed chemobrain pathologies as indicated by brain inflammatory and oxidative insults, glial activation, defective mitochondrial homeostasis, increased potential lesions associated with Alzheimer's disease, disrupted neurogenesis, loss of dendritic spines, and ultimately neuronal death through both apoptosis and necroptosis. Intervention with DPZ co-treatment completely restored cognitive function by attenuating these pathological conditions induced by DOX. We also confirmed that DPZ treatment does not affect the anti-cancer efficacy of Dox in breast cancer cells. Together, our findings suggest that DPZ treatment confers potential neuroprotection against Dox-induced chemobrain.
Collapse
Affiliation(s)
- Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Thawatchai Khuanjing
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Patcharapong Pantiya
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Neuroelectrophysiology Unit, Chiang Mai University, 50200, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, 50200, Chiang Mai, Thailand.
| |
Collapse
|
27
|
Ng Kee Kwong KC, Harbham PK, Selvaraj BT, Gregory JM, Pal S, Hardingham GE, Chandran S, Mehta AR. 40 Years of CSF Toxicity Studies in ALS: What Have We Learnt About ALS Pathophysiology? Front Mol Neurosci 2021; 14:647895. [PMID: 33815058 PMCID: PMC8012723 DOI: 10.3389/fnmol.2021.647895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Based on early evidence of in vitro neurotoxicity following exposure to serum derived from patients with amyotrophic lateral sclerosis (ALS), several studies have attempted to explore whether cerebrospinal fluid (CSF) obtained from people with ALS could possess similar properties. Although initial findings proved inconclusive, it is now increasingly recognized that ALS-CSF may exert toxicity both in vitro and in vivo. Nevertheless, the mechanism underlying CSF-induced neurodegeneration remains unclear. This review aims to summarize the 40-year long history of CSF toxicity studies in ALS, while discussing the various mechanisms that have been proposed, including glutamate excitotoxicity, proteotoxicity and oxidative stress. Furthermore, we consider the potential implications of a toxic CSF circulatory system in the pathophysiology of ALS, and also assess its significance in the context of current ALS research.
Collapse
Affiliation(s)
| | - Pratap K. Harbham
- West Midlands Academic Foundation Programme, University of Birmingham, Birmingham, United Kingdom
| | - Bhuvaneish T. Selvaraj
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna M. Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- MRC Edinburgh Brain Bank, Academic Department of Neuropathology, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Pathology, University of Edinburgh, Edinburgh, United Kingdom
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E. Hardingham
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Development and Repair, InStem, Bengaluru, India
| | - Arpan R. Mehta
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, United Kingdom
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|