1
|
Kemppainen S, Aramburu-Núñez M, Koivisto H, Posado-Fernández A, Felix-Garcia MR, Nurminen K, Häkli S, Abelli-Deulofeu E, Kaisto M, Custodia A, van Olst L, Willman RM, Mäkinen P, Miettinen PO, Schouten M, Iglesias-Rey R, Kamermans A, Martiskainen H, Rauramaa T, de Vries HE, Eng E, Salo R, Kettunen M, Gröhn O, Gureviciene I, Chmielevski P, Ouro A, Lapresa R, Agulla J, Almeida A, Doria G, Flores O, Aguiar P, Carmans S, Roucourt B, Hiltunen M, Sobrino T, Tanila H. Intracerebroventricular phospho-tau immunotherapy alleviates cortical phospho-tau burden and motor phenotype in a neuron-specific P301S tauopathy mouse. Exp Neurol 2025; 391:115315. [PMID: 40403829 DOI: 10.1016/j.expneurol.2025.115315] [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: 01/27/2025] [Revised: 05/11/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Pathological tau accumulation disrupts neuronal function, leading to neurodegeneration and dementia in tauopathies, such as Alzheimer's disease and frontotemporal dementia. Despite the progression of several anti-tau therapies to clinical trials, no disease-modifying treatments for tauopathies exist. Tau hyperphosphorylation is a key factor in pathology progression. Among all tau phosphorylation sites targeted in preclinical passive immunization studies, the classic AT8 pathological tau phosphorylation sites have remained understudied. Thus, we investigated the potential of immunotherapy against phosphorylated tau (pTau) in a P301S mouse model of tauopathy. We administered a new monoclonal B6 antibody that targets tau phosphorylation sites at serine 202, threonine 205 and serine 208 either systemically for 3 months or locally into the cerebral ventricles for 1 or 2 months via an osmotic minipump. Systemic administration failed to reach the brain effectively, and subsequently, was not able to alleviate the progressive motor impairment seen in this tauopathy mouse model. By contrast, intraventricular administration improved motor function in earlier stages of pathology but had a lesser effect in later stages. The local administration for 8 weeks reduced the number of pTau positive neurons in cortex and hippocampus. Our findings indicate that targeting the classical pathological tau phosphorylation sites can ameliorate tau pathology and improve function in a mouse tauopathy model. These results add to growing evidence supporting the efforts in developing tau-targeting immunotherapies for neurodegenerative diseases associated with pathological tau deposits.
Collapse
Affiliation(s)
- S Kemppainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - M Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - H Koivisto
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - A Posado-Fernández
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; STAB VIDA Lda, Caparica, Portugal
| | - M R Felix-Garcia
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - K Nurminen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - S Häkli
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - E Abelli-Deulofeu
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - M Kaisto
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - A Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - L van Olst
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - R-M Willman
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - P Mäkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - P O Miettinen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - M Schouten
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - A Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands
| | - H Martiskainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - T Rauramaa
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland; Institute of Clinical Medicine-Pathology, University of Eastern Finland, Kuopio, Finland
| | - H E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands; Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - E Eng
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - R Salo
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - M Kettunen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - O Gröhn
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - I Gureviciene
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | - A Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - R Lapresa
- Institute of Functional Biology and Genomics, CSIC, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, Salamanca, Spain
| | - J Agulla
- Institute of Functional Biology and Genomics, CSIC, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, Salamanca, Spain
| | - A Almeida
- Institute of Functional Biology and Genomics, CSIC, Universidad de Salamanca, Salamanca, Spain; Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, Salamanca, Spain
| | - G Doria
- STAB VIDA Lda, Caparica, Portugal
| | - O Flores
- STAB VIDA Lda, Caparica, Portugal
| | - P Aguiar
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | | | | | - M Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - T Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - H Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
2
|
Berlind JE, Lai JD, Lie C, Vicente J, Lam K, Guo S, Chang J, Yu V, Ichida JK. KCTD20 suppression mitigates excitotoxicity in tauopathy patient organoids. Neuron 2025; 113:1169-1189.e7. [PMID: 40049159 PMCID: PMC12005969 DOI: 10.1016/j.neuron.2025.02.001] [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: 05/07/2024] [Revised: 12/13/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025]
Abstract
Excitotoxicity is a major pathologic mechanism in patients with tauopathy and other neurodegenerative diseases. However, the key neurotoxic drivers and the most effective strategies for mitigating these degenerative processes are unclear. Here, we show that glutamate treatment of induced pluripotent stem cell (iPSC)-derived cerebral organoids induces tau oligomerization and neurodegeneration and that these phenotypes are enhanced in organoids derived from tauopathy patients. Using a genome-wide CRISPR interference (CRISPRi) screen, we find that the suppression of KCTD20 potently ameliorates tau pathology and neurodegeneration in glutamate-treated organoids and mice, as well as in transgenic mice overexpressing mutant human tau. KCTD20 suppression reduces oligomeric tau and improves neuron survival by activating lysosomal exocytosis, which clears pathological tau. Our results show that glutamate signaling can induce neuronal tau pathology and identify KCTD20 suppression and lysosomal exocytosis as effective strategies for clearing neurotoxic tau species.
Collapse
Affiliation(s)
- Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA; Department of Neuroscience, Amgen Inc., Cambridge, MA, USA; Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA.
| | - Cecilia Lie
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jokabeth Vicente
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Kelsey Lam
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Sheron Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Jonathan Chang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA
| | - Violeta Yu
- Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Böken D, Wu Y, Zhang Z, Klenerman D. Detecting the Undetectable: Advances in Methods for Identifying Small Tau Aggregates in Neurodegenerative Diseases. Chembiochem 2025; 26:e202400877. [PMID: 39688878 PMCID: PMC12002113 DOI: 10.1002/cbic.202400877] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/18/2024]
Abstract
Tau, a microtubule-associated protein, plays a critical role in maintaining neuronal structure and function. However, in neurodegenerative diseases such as Alzheimer's disease and other tauopathies, tau misfolds and aggregates into oligomers and fibrils, leading to neuronal damage. Tau oligomers are increasingly recognised as the most neurotoxic species, inducing synaptic dysfunction and contributing to disease progression. Detecting these early-stage aggregates is challenging due to their low concentration and high heterogeneity in biological samples. Traditional methods such as immunostaining and enzyme-linked immunosorbent assay (ELISA) lack the sensitivity and specificity to reliably detect small tau aggregates. Advanced single-molecule approaches, including single-molecule fluorescence resonance energy transfer (smFRET) and single-molecule pull-down (SiMPull), offer improved sensitivity for studying tau aggregation at the molecular level. These emerging tools provide critical insights into tau pathology, enabling earlier detection and characterisation of disease-relevant aggregates, thereby offering potential for the development of targeted therapies and diagnostic approaches for tauopathies.
Collapse
Affiliation(s)
- Dorothea Böken
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeCB2 0AHUK
| | - Yunzhao Wu
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeCB2 0AHUK
| | - Ziwei Zhang
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeCB2 0AHUK
| | - David Klenerman
- Yusuf Hamied Department of ChemistryUniversity of CambridgeCambridgeCB2 1EWUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeCB2 0AHUK
| |
Collapse
|
4
|
Mohamed AS, ElKaffas M, Metwally K, Abdelfattah M, Elsery EA, Elshazly A, Gomaa HE, Alsayed A, El-Desouky S, El-Gamal R, Elfarrash S. Impairment of Nrf2 signaling in the hippocampus of P301S tauopathy mice model aligns with the cognitive impairment and the associated neuroinflammation. J Inflamm (Lond) 2024; 21:29. [PMID: 39107774 PMCID: PMC11304845 DOI: 10.1186/s12950-024-00396-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/24/2024] [Indexed: 08/10/2024] Open
Abstract
Mice transgenic for human P301S tau protein exhibit many characteristics of the human tauopathies, including the formation of abundant hyperphoshorylated tau filaments, the associated neuroinflammation and disease phenotype. However, the exact underpinning mechanisms are still not fully addressed that hinder our understanding of the tauopathy diseases and the development of possible therapeutic targets.Methods: In the current study, hippocampus from three disease time points (2, 4 and 6 months) of P301S mice were further characterized in comparison to the age and sex matched control wild type mice (WT) that do not express the transgene. Different spectrum of hippocampal dependent cognitive tests, biochemical and pathological analysis were conducted to understand the disease progression and the associated changes in each stage. Results: Cognitive impairment was manifested as early as 2 months age, prior to the identification of tau aggregation and phosphorylation by immunostaining. P301S mice manifested an increased pro-inflammatory related changes at mRNA transcription level (IL-1b and IL17A) with the progression of the disease and when compared to the WT mice of the same age. Among the identified genes in the current study, the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) genes expression that is considered as the master regulator of an endogenous inducible defense system was significantly impaired in P301S mice by 4 and 6 months when compared to healthy WT controls. A data that was also supported by the immunostaining of the serial brain sections including the both brain stem and hippocampus. The current result is suggesting that the downregulation of Nrf2 gene and the impaired Nrf2 dependent anti-inflammatory mechanisms in P301S mice brain is possibly contributing -among other factors- in the neuroinflammation and tauopathy, and that modulation of Nrf2 signaling impairments can be further investigated as a promising potential therapeutic target for tauopathy.
Collapse
Affiliation(s)
- Ahmed Sabry Mohamed
- Program of Medicine, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Mahmoud ElKaffas
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Karim Metwally
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Mahmoud Abdelfattah
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Eslam Ashraf Elsery
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed Elshazly
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Hossam Eldin Gomaa
- Mansoura Manchester Medical Program, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Aziza Alsayed
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Sara El-Desouky
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Randa El-Gamal
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
- Department of Medical Biochemistry, Faculty of Medicine, Horus University, New Damietta, Egypt
- Department of Medical Biochemistry, Faculty of Medicine, New Mansoura University, Mansoura, Egypt
| | - Sara Elfarrash
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
5
|
Sidoryk-Węgrzynowicz M, Adamiak K, Strużyńska L. Astrocyte-Neuron Interaction via the Glutamate-Glutamine Cycle and Its Dysfunction in Tau-Dependent Neurodegeneration. Int J Mol Sci 2024; 25:3050. [PMID: 38474295 DOI: 10.3390/ijms25053050] [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: 02/14/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Astroglia constitute the largest group of glial cells and are involved in numerous actions that are critical to neuronal development and functioning, such as maintaining the blood-brain barrier, forming synapses, supporting neurons with nutrients and trophic factors, and protecting them from injury. These properties are deeply affected in the course of many neurodegenerative diseases, including tauopathies, often before the onset of the disease. In this respect, the transfer of essential amino acids such as glutamate and glutamine between neurons and astrocytes in the glutamate-glutamine cycle (GGC) is one example. In this review, we focus on the GGC and the disruption of this cycle in tau-dependent neurodegeneration. A profound understanding of the complex functions of the GGC and, in the broader context, searching for dysfunctions in communication pathways between astrocytes and neurons via GGC in health and disease, is of critical significance for the development of novel mechanism-based therapies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Marta Sidoryk-Węgrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland
| | - Kamil Adamiak
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland
| |
Collapse
|
6
|
Eisenbaum M, Pearson A, Ortiz C, Mullan M, Crawford F, Ojo J, Bachmeier C. ApoE4 expression disrupts tau uptake, trafficking, and clearance in astrocytes. Glia 2024; 72:184-205. [PMID: 37668005 DOI: 10.1002/glia.24469] [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: 02/03/2023] [Revised: 08/07/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
Tauopathies are a collection of neurodegenerative diseases characterized by the accumulation of pathogenic aggregates of the microtubule-associated protein tau. Despite the prevalence and diversity of tau astrogliopathy in tauopathies, the interactions between astrocytes and tau in the brain, and the influence of neurodegenerative genetic risk factors like the apolipoprotein E4 (apoE4) isoform, are largely unknown. Here, we leveraged primary and immortalized astrocytes expressing humanized apoE isoforms to characterize the mechanisms by which astrocytes interact with and eliminate extracellular tau, and the influence of apoE genotype on these processes. Our work indicates that astrocytes rapidly internalize, process, and release tau via an exosomal secretory mechanism under physiological conditions. However, we found that apoE4 disrupted these processes in comparison to apoE3, resulting in an astrocytic phenotype prone to intracellular tau accumulation. Furthermore, exposure to repetitive mild traumatic brain injuries exacerbated the apoE4-induced impairments in tau processing and elimination by astrocytes in apoE4 targeted-replacement mice. The diminished ability of apoE4 astrocytes to eliminate extracellular tau can lead to an accumulation of pathogenic tau, which induces mitochondrial dysfunction, as demonstrated by our studies. In total, our findings suggest that the apoE4 isoform lowers the threshold of astrocytic resilience to pathogenic tau, rendering them susceptible to bioenergetic deficits in the early stages of neurodegenerative diseases such as traumatic brain injury, potentially contributing to neurological decline.
Collapse
Affiliation(s)
| | | | | | | | - Fiona Crawford
- The Roskamp Institute, Sarasota, Florida, USA
- James A. Haley Veterans' Hospital, Tampa, Florida, USA
| | - Joseph Ojo
- The Roskamp Institute, Sarasota, Florida, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, Florida, USA
- Bay Pines VA Healthcare System, Bay Pines, Florida, USA
| |
Collapse
|
7
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
8
|
Chen H, Fan L, Guo Q, Wong MY, Yu F, Foxe N, Wang W, Nessim A, Carling G, Liu B, Lopez-Lee C, Huang Y, Amin S, Patel T, Mok SA, Song WM, Zhang B, Ma Q, Fu H, Gan L, Luo W. DAP12 deficiency alters microglia-oligodendrocyte communication and enhances resilience against tau toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.563970. [PMID: 37961594 PMCID: PMC10634844 DOI: 10.1101/2023.10.26.563970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.
Collapse
Affiliation(s)
- Hao Chen
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qi Guo
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nessa Foxe
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Aviram Nessim
- The State University of New York at Stony Brook, Long Island, New York, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Tark Patel
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Sue-Ann Mok
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Millburn High School, New Jersey, NJ, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
9
|
Chen H, Fan L, Guo Q, Wong MY, Yu F, Foxe N, Wang W, Nessim A, Carling G, Liu B, Lopez-Lee C, Huang Y, Amin S, Mok SA, Song WM, Zhang B, Ma Q, Fu H, Gan L, Luo W. DAP12 deficiency alters microglia-oligodendrocyte communication and enhances resilience against tau toxicity. RESEARCH SQUARE 2023:rs.3.rs-3454358. [PMID: 37961627 PMCID: PMC10635319 DOI: 10.21203/rs.3.rs-3454358/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.
Collapse
Affiliation(s)
- Hao Chen
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qi Guo
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nessa Foxe
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Aviram Nessim
- The State University of New York at Stony Brook, Long Island, New York, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sue-Ann Mok
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Millburn High School, New Jersey, NJ, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Nutma E, Fancy N, Weinert M, Tsartsalis S, Marzin MC, Muirhead RCJ, Falk I, Breur M, de Bruin J, Hollaus D, Pieterman R, Anink J, Story D, Chandran S, Tang J, Trolese MC, Saito T, Saido TC, Wiltshire KH, Beltran-Lobo P, Phillips A, Antel J, Healy L, Dorion MF, Galloway DA, Benoit RY, Amossé Q, Ceyzériat K, Badina AM, Kövari E, Bendotti C, Aronica E, Radulescu CI, Wong JH, Barron AM, Smith AM, Barnes SJ, Hampton DW, van der Valk P, Jacobson S, Howell OW, Baker D, Kipp M, Kaddatz H, Tournier BB, Millet P, Matthews PM, Moore CS, Amor S, Owen DR. Translocator protein is a marker of activated microglia in rodent models but not human neurodegenerative diseases. Nat Commun 2023; 14:5247. [PMID: 37640701 PMCID: PMC10462763 DOI: 10.1038/s41467-023-40937-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Microglial activation plays central roles in neuroinflammatory and neurodegenerative diseases. Positron emission tomography (PET) targeting 18 kDa Translocator Protein (TSPO) is widely used for localising inflammation in vivo, but its quantitative interpretation remains uncertain. We show that TSPO expression increases in activated microglia in mouse brain disease models but does not change in a non-human primate disease model or in common neurodegenerative and neuroinflammatory human diseases. We describe genetic divergence in the TSPO gene promoter, consistent with the hypothesis that the increase in TSPO expression in activated myeloid cells depends on the transcription factor AP1 and is unique to a subset of rodent species within the Muroidea superfamily. Finally, we identify LCP2 and TFEC as potential markers of microglial activation in humans. These data emphasise that TSPO expression in human myeloid cells is related to different phenomena than in mice, and that TSPO-PET signals in humans reflect the density of inflammatory cells rather than activation state.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Nurun Fancy
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria Weinert
- Department of Brain Sciences, Imperial College London, London, UK
| | - Stergios Tsartsalis
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Manuel C Marzin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robert C J Muirhead
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Irene Falk
- Viral Immunology Section, NIH, Bethesda, MD, USA
- Flow and Imaging Cytometry Core Facility, NIH, Bethesda, MD, USA
| | - Marjolein Breur
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Joy de Bruin
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Robin Pieterman
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | - Jasper Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - David Story
- UK Dementia Research Institute at Edinburgh, Edinburgh, UK
| | | | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria C Trolese
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Takaomi C Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Nagoya, Japan
| | | | - Paula Beltran-Lobo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alexandra Phillips
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Luke Healy
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Marie-France Dorion
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Dylan A Galloway
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Rochelle Y Benoit
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Quentin Amossé
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Kelly Ceyzériat
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | | | - Enikö Kövari
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Caterina Bendotti
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Carola I Radulescu
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Jia Hui Wong
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Anna M Barron
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Amy M Smith
- UK Dementia Research Institute at Imperial College London, London, UK
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Samuel J Barnes
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | | | - Paul van der Valk
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands
| | | | - Owain W Howell
- Institute of Life Science (ILS), Swansea University Medical School, Swansea, UK
| | - David Baker
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | - Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany
| | | | - Philippe Millet
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
- Division of Adult Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Craig S Moore
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, Canada
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC - Location VUmc, Amsterdam, The Netherlands.
- Department of Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, UK.
- Institute of Anatomy, Rostock University Medical Center, 18057, Rostock, Germany.
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at Imperial College London, London, UK.
| |
Collapse
|
11
|
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
|
12
|
Kunze LH, Ruch F, Biechele G, Eckenweber F, Wind-Mark K, Dinkel L, Feyen P, Bartenstein P, Ziegler S, Paeger L, Tahirovic S, Herms J, Brendel M. Long-Term Pioglitazone Treatment Has No Significant Impact on Microglial Activation and Tau Pathology in P301S Mice. Int J Mol Sci 2023; 24:10106. [PMID: 37373253 DOI: 10.3390/ijms241210106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Neuroinflammation is one disease hallmark on the road to neurodegeneration in primary tauopathies. Thus, immunomodulation might be a suitable treatment strategy to delay or even prevent the occurrence of symptoms and thus relieve the burden for patients and caregivers. In recent years, the peroxisome proliferator-activated receptor γ (PPARγ) has received increasing attention as it is immediately involved in the regulation of the immune system and can be targeted by the anti-diabetic drug pioglitazone. Previous studies have shown significant immunomodulation in amyloid-β (Aβ) mouse models by pioglitazone. In this study, we performed long-term treatment over six months in P301S mice as a tauopathy model with either pioglitazone or placebo. We performed serial 18 kDa translocator protein positron-emission-tomography (TSPO-PET) imaging and terminal immunohistochemistry to assess microglial activation during treatment. Tau pathology was quantified via immunohistochemistry at the end of the study. Long-term pioglitazone treatment had no significant effect on TSPO-PET, immunohistochemistry read-outs of microglial activation, or tau pathology levels in P301S mice. Thus, we conclude that pioglitazone modifies the time course of Aβ-dependent microglial activation, but does not significantly modulate microglial activation in response to tau pathology.
Collapse
Affiliation(s)
- Lea Helena Kunze
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - François Ruch
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Gloria Biechele
- Department of Radiology, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Lina Dinkel
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Paul Feyen
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Lars Paeger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University of Munich, 81377 Munich, Germany
- Center for Neuropathology and Prion Research, LMU Munich, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Feodor-Lynen-Str. 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University of Munich, 81377 Munich, Germany
| |
Collapse
|
13
|
Sidoryk-Węgrzynowicz M, Dąbrowska-Bouta B, Sulkowski G, Strużyńska L. Mutant Tau protein-induced abnormalities in the Na +-dependent glutamine translocation and recycling and their impact on astrocyte-neuron integrity in vitro. Neurochem Int 2023; 168:105551. [PMID: 37295680 DOI: 10.1016/j.neuint.2023.105551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
Tau-dependent neurodegeneration is accompanied by astrocytosis in a mouse trans-genic model, which replicates the neuropathological characteristic of tauopathy and other human neurodegenerative disorders where astrocyte activation precedes neuronal loss and is associated with disease progression. This indicates an important role of astrocytes in the development of the disease. Astrocytes derived from a transgenic mouse model expressing human Tau, exhibit changes in cellular markers of astrocyte neuroprotective function related to the glutamate-glutamine cycle (GGC), representing a key part of astrocyte-neuron integrity. Here, we focused on investigating the functional properties of key GGC components involved in the astrocyte-neuron network associated with Tau pathology in vitro. Mutant recombinant Tau (rTau) carrying the P301L mutation was added to the neuronal cultures, with or without control astrocyte-conditioned medium (ACM), to study glutamine translocation through the GGC. We demonstrated that mutant Tau in vitro induces neuronal degeneration, while control astrocytes response in neuroprotective way by preventing neurodegeneration. In parallel with this observation, we noticed the Tau-dependent decline of neuronal microtubule associated protein 2 (MAP2), followed by changes in glutamine (Gln) transport. Exposure to rTau decreases sodium-dependent Gln uptake in neurons and that effect was reversed when cells were co-incubated with control ACM after induction of rTau dependent pathology. Further, we found that neuronal Na+-dependent system A is the most specific system that is affected under rTau exposure. In addition, in rTau-treated astrocytes total Na+-dependent uptake of Gln, which is mediated by the N system, increases. Altogether, our study suggest mechanisms operating in Tau pathology may be related to the alterations in glutamine transport and recycling that affect neuronal-astrocytic integrity.
Collapse
Affiliation(s)
- Marta Sidoryk-Węgrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str, 02-106, Warsaw, Poland.
| | - Beata Dąbrowska-Bouta
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str, 02-106, Warsaw, Poland
| | - Grzegorz Sulkowski
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str, 02-106, Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str, 02-106, Warsaw, Poland
| |
Collapse
|
14
|
Kemppainen S, Huber N, Willman RM, Zamora A, Mäkinen P, Martiskainen H, Takalo M, Haapasalo A, Sobrino T, González Gómez MA, Piñeiro Y, Rivas J, Himmelreich U, Hiltunen M. Organotypic Hippocampal Slice Cultures from Adult Tauopathy Mice and Theragnostic Evaluation of Nanomaterial Phospho-TAU Antibody-Conjugates. Cells 2023; 12:1422. [PMID: 37408256 DOI: 10.3390/cells12101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Organotypic slice culture models surpass conventional in vitro methods in many aspects. They retain all tissue-resident cell types and tissue hierarchy. For studying multifactorial neurodegenerative diseases such as tauopathies, it is crucial to maintain cellular crosstalk in an accessible model system. Organotypic slice cultures from postnatal tissue are an established research tool, but adult tissue-originating systems are missing, yet necessary, as young tissue-originating systems cannot fully model adult or senescent brains. To establish an adult-originating slice culture system for tauopathy studies, we made hippocampal slice cultures from transgenic 5-month-old hTau.P301S mice. In addition to the comprehensive characterization, we set out to test a novel antibody for hyperphosphorylated TAU (pTAU, B6), with and without a nanomaterial conjugate. Adult hippocampal slices retained intact hippocampal layers, astrocytes, and functional microglia during culturing. The P301S-slice neurons expressed pTAU throughout the granular cell layer and secreted pTAU to the culture medium, whereas the wildtype slices did not. Additionally, cytotoxicity and inflammation-related determinants were increased in the P301S slices. Using fluorescence microscopy, we showed target engagement of the B6 antibody to pTAU-expressing neurons and a subtle but consistent decrease in intracellular pTAU with the B6 treatment. Collectively, this tauopathy slice culture model enables measuring the extracellular and intracellular effects of different mechanistic or therapeutic manipulations on TAU pathology in adult tissue without the hindrance of the blood-brain barrier.
Collapse
Affiliation(s)
- Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Nadine Huber
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Roosa-Maria Willman
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ana Zamora
- Molecular Imaging and Photonics, KU Leuven, 3001 Leuven, Belgium
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Manuel Antonio González Gómez
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Yolanda Piñeiro
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Rivas
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
15
|
Early impairments of visually-driven neuronal ensemble dynamics in the rTg4510 tauopathy mouse model. Neurobiol Dis 2023; 178:106012. [PMID: 36696792 DOI: 10.1016/j.nbd.2023.106012] [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: 10/10/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023] Open
Abstract
Tau protein pathology is a hallmark of many neurodegenerative diseases, including Alzheimer's Disease or frontotemporal dementia. Synaptic dysfunction and abnormal visual evoked potentials have been reported in murine models of tauopathy, but little is known about the state of the network activity on a single neuronal level prior to brain atrophy. In the present study, oscillatory rhythms and single-cell calcium activity of primary visual cortex pyramidal neuron population were investigated in basal and light evoked states in the rTg4510 tauopathy mouse model prior to neurodegeneration. We found a decrease in their responsivity and overall activity which was insensitive to GABAergic modulation. Despite an enhancement of basal state coactivation of cortical pyramidal neurons, a loss of input-output synchronicity was observed. Dysfunction of cortical pyramidal function was also reflected in a reduction of basal theta oscillations and enhanced susceptibility to a sub-convulsive dose of pentylenetetrazol in rTg4510 mice. Our results unveil impairments in visual cortical pyramidal neuron processing and define aberrant oscillations as biomarker candidates in early stages of neurodegenerative tauopathies.
Collapse
|
16
|
Wenger K, Viode A, Schlaffner CN, van Zalm P, Cheng L, Dellovade T, Langlois X, Bannon A, Chang R, Connors TR, Oakley D, Renard B, Rappsilber J, Hyman B, Steen H, Steen JA. Common mouse models of tauopathy reflect early but not late human disease. Mol Neurodegener 2023; 18:10. [PMID: 36732784 PMCID: PMC9893608 DOI: 10.1186/s13024-023-00601-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Mouse models that overexpress human mutant Tau (P301S and P301L) are commonly used in preclinical studies of Alzheimer's Disease (AD) and while several drugs showed therapeutic effects in these mice, they were ineffective in humans. This leads to the question to which extent the murine models reflect human Tau pathology on the molecular level. METHODS We isolated insoluble, aggregated Tau species from two common AD mouse models during different stages of disease and characterized the modification landscape of the aggregated Tau using targeted and untargeted mass spectrometry-based proteomics. The results were compared to human AD and to human patients that suffered from early onset dementia and that carry the P301L Tau mutation. RESULTS Both mouse models accumulate insoluble Tau species during disease. The Tau aggregation is driven by progressive phosphorylation within the proline rich domain and the C-terminus of the protein. This is reflective of early disease stages of human AD and of the pathology of dementia patients carrying the P301L Tau mutation. However, Tau ubiquitination and acetylation, which are important to late-stage human AD are not represented in the mouse models. CONCLUSION AD mouse models that overexpress human Tau using risk mutations are a suitable tool for testing drug candidates that aim to intervene in the early formation of insoluble Tau species promoted by increased phosphorylation of Tau.
Collapse
Affiliation(s)
- Kathrin Wenger
- F.M. Kirby Neurobiology Center, Department of Neurobiology, Boston Children's Hospital and Harvard, Medical School; Center for Life Science, RM 12030, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Arthur Viode
- Departments of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Christoph N Schlaffner
- Departments of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Data Analytics and Computational Statistics, Hasso-Plattner-Institute, Faculty of Digital Engineering; University of Potsdam, Potsdam, Germany
| | - Patrick van Zalm
- Departments of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Long Cheng
- F.M. Kirby Neurobiology Center, Department of Neurobiology, Boston Children's Hospital and Harvard, Medical School; Center for Life Science, RM 12030, 3 Blackfan Circle, Boston, MA 02115, USA
| | | | | | | | - Rui Chang
- AbbVie, Cambridge Research Center, Cambridge, MA, USA
| | - Theresa R Connors
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Derek Oakley
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bernhard Renard
- Data Analytics and Computational Statistics, Hasso-Plattner-Institute, Faculty of Digital Engineering; University of Potsdam, Potsdam, Germany
| | - Juri Rappsilber
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Bradley Hyman
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hanno Steen
- Departments of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Neurobiology Program, Boston Children's Hospital, Boston, MA, USA
| | - Judith A Steen
- F.M. Kirby Neurobiology Center, Department of Neurobiology, Boston Children's Hospital and Harvard, Medical School; Center for Life Science, RM 12030, 3 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
17
|
CSF1R inhibitors induce a sex-specific resilient microglial phenotype and functional rescue in a tauopathy mouse model. Nat Commun 2023; 14:118. [PMID: 36624100 PMCID: PMC9829908 DOI: 10.1038/s41467-022-35753-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.
Collapse
|
18
|
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
|
19
|
Tong BCK, Huang AS, Wu AJ, Iyaswamy A, Ho OKY, Kong AHY, Sreenivasmurthy SG, Zhu Z, Su C, Liu J, Song J, Li M, Cheung KH. Tetrandrine ameliorates cognitive deficits and mitigates tau aggregation in cell and animal models of tauopathies. J Biomed Sci 2022; 29:85. [PMID: 36273169 PMCID: PMC9587578 DOI: 10.1186/s12929-022-00871-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/19/2022] [Indexed: 12/05/2022] Open
Abstract
Background Tauopathies are neurodegenerative diseases that are associated with the pathological accumulation of tau-containing tangles in the brain. Tauopathy can impair cognitive and motor functions and has been observed in Alzheimer’s disease (AD) and frontotemporal dementia (FTD). The aetiology of tauopathy remains mysterious; however, recent studies suggest that the autophagic-endolysosomal function plays an essential role in the degradation and transmission of pathological tau. We previously demonstrated that tetrandrine could ameliorate memory functions and clear amyloid plaques in transgenic AD mice by restoring autophagic-endolysosomal function. However, the efficacy of tetrandrine and the associated therapeutic mechanism in tauopathies have not been evaluated and elucidated. Methods Novel object recognition, fear conditioning and electrophysiology were used to evaluate the effects of tetrandrine on memory functions in transgenic tau mice. Western blotting and immunofluorescence staining were employed to determine the effect of tetrandrine on autophagy and tau clearance in vivo. Calcium (Ca2+) imaging and flow cytometry were used to delineate the role of pathological tau and tetrandrine in lysosomal Ca2+ and pH homeostasis. Biochemical BiFC fluorescence, Western blotting and immunofluorescence staining were used to evaluate degradation of hyperphosphorylated tau in vitro, whereas coculture of brain slices with isolated microglia was used to evaluate tau clearance ex vivo. Results We observed that tetrandrine treatment mitigated tau tangle development and corrected memory impairment in Thy1-hTau.P301S transgenic mice. Mechanistically, we showed that mutant tau expression disrupts lysosome pH by increasing two-pore channel 2 (TPC2)-mediated Ca2+ release, thereby contributing to lysosome alkalinization. Tetrandrine inhibits TPC2, thereby restoring the lysosomal pH, promotes tau degradation via autophagy, and ameliorates tau aggregation. Furthermore, in an ex vivo assay, we demonstrated that tetrandrine treatment promotes pathological tau clearance by microglia. Conclusions Together, these findings suggest that pathological tau disturbs endolysosomal homeostasis to impair tau clearance. This impairment results in a vicious cycle that accelerates disease pathogenesis. The success of tetrandrine in reducing tau aggregation suggests first, that tetrandrine could be an effective drug for tauopathies and second, that rescuing lysosomal Ca2+ homeostasis, thereby restoring ALP function, could be an effective general strategy for the development of novel therapies for tauopathies. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00871-6.
Collapse
Affiliation(s)
- Benjamin Chun-Kit Tong
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China.
| | - Alexis Shiying Huang
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Aston Jiaxi Wu
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Ashok Iyaswamy
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Olivia Ka-Yi Ho
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Anna Hau-Yee Kong
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Sravan Gopalkrishnashetty Sreenivasmurthy
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Zhou Zhu
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Chengfu Su
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Jia Liu
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China
| | - Juxian Song
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Li
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China.
| | - King-Ho Cheung
- School of Chinese Medicine and Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, SAR, China.
| |
Collapse
|
20
|
Feng S, Zhang T, Ke W, Xiao Y, Guo Z, Lu C, Li S, Guo Z, Liu Y, Tan G, Chen Y, Yue F, Shu Y, Yue C, Jing N. The long-term survival and functional maturation of human iNPC-derived neurons in the basal forebrain of cynomolgus monkeys. LIFE MEDICINE 2022; 1:196-206. [PMID: 39871935 PMCID: PMC11749281 DOI: 10.1093/lifemedi/lnac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/27/2022] [Indexed: 01/29/2025]
Abstract
Human induced neural stem/progenitor cells (iNPCs) are a promising source of cells for stem cell-based therapy. The therapeutic potential of human iNPCs has been extensively tested in animal models, including both mouse and monkey models. However, the comprehensive characterization of grafted iNPCs in the brain of non-human primates has been lagged behind. In this study, we transplanted human iNPCs into the basal forebrain of adult cynomolgus monkeys. We found that grafted iNPCs predominantly differentiated into neurons that displayed long-term survival up to 12 months. Additionally, iNPC-derived human neurons gradually matured in term of morphology and subtype differentiation. More excitingly, we observed that human neurons displayed electrophysiological activities resembling those of mature neurons, indicating the acquisition of functional membrane properties. Collectively, this study systematically characterized human iNPCs in the brain of non-human primates, and will provide invaluable clues for developing safe and effective stem cell-based therapies for different brain disorders.
Collapse
Affiliation(s)
- Su Feng
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou 510005, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Wei Ke
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yujie Xiao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zhong Guo
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
| | - Chunling Lu
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
| | - Shuntang Li
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhongxin Guo
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yuanyuan Liu
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Guohe Tan
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Feng Yue
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Yousheng Shu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Department of Biological Sciences, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou 215000, China
| | - Naihe Jing
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou 510005, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
21
|
Bold CS, Baltissen D, Ludewig S, Back MK, Just J, Kilian L, Erdinger S, Banicevic M, Rehra L, Almouhanna F, Nigri M, Wolfer DP, Spilger R, Rohr K, Kann O, Buchholz CJ, von Engelhardt J, Korte M, Müller UC. APPsα Rescues Tau-Induced Synaptic Pathology. J Neurosci 2022; 42:5782-5802. [PMID: 35667850 PMCID: PMC9302470 DOI: 10.1523/jneurosci.2200-21.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/06/2022] [Accepted: 05/22/2022] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is histopathologically characterized by Aβ plaques and the accumulation of hyperphosphorylated Tau species, the latter also constituting key hallmarks of primary tauopathies. Whereas Aβ is produced by amyloidogenic APP processing, APP processing along the competing nonamyloidogenic pathway results in the secretion of neurotrophic and synaptotrophic APPsα. Recently, we demonstrated that APPsα has therapeutic effects in transgenic AD model mice and rescues Aβ-dependent impairments. Here, we examined the potential of APPsα to mitigate Tau-induced synaptic deficits in P301S mice (both sexes), a widely used mouse model of tauopathy. Analysis of synaptic plasticity revealed an aberrantly increased LTP in P301S mice that could be normalized by acute application of nanomolar amounts of APPsα to hippocampal slices, indicating a homeostatic function of APPsα on a rapid time scale. Further, AAV-mediated in vivo expression of APPsα restored normal spine density of CA1 neurons even at stages of advanced Tau pathology not only in P301S mice, but also in independent THY-Tau22 mice. Strikingly, when searching for the mechanism underlying aberrantly increased LTP in P301S mice, we identified an early and progressive loss of major GABAergic interneuron subtypes in the hippocampus of P301S mice, which may lead to reduced GABAergic inhibition of principal cells. Interneuron loss was paralleled by deficits in nest building, an innate behavior highly sensitive to hippocampal impairments. Together, our findings indicate that APPsα has therapeutic potential for Tau-mediated synaptic dysfunction and suggest that loss of interneurons leads to disturbed neuronal circuits that compromise synaptic plasticity as well as behavior.SIGNIFICANCE STATEMENT Our findings indicate, for the first time, that APPsα has the potential to rescue Tau-induced spine loss and abnormal synaptic plasticity. Thus, APPsα might have therapeutic potential not only because of its synaptotrophic functions, but also its homeostatic capacity for neuronal network activity. Hence, APPsα is one of the few molecules which has proven therapeutic effects in mice, both for Aβ- and Tau-dependent synaptic impairments and might therefore have therapeutic potential for patients suffering from AD or primary tauopathies. Furthermore, we found in P301S mice a pronounced reduction of inhibitory interneurons as the earliest pathologic event preceding the accumulation of hyperphosphorylated Tau species. This loss of interneurons most likely disturbs neuronal circuits that are important for synaptic plasticity and behavior.
Collapse
Affiliation(s)
- Charlotte S Bold
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Danny Baltissen
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Susann Ludewig
- TU Braunschweig, Zoological Institute, Braunschweig, 38106, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, 38124, Germany
| | - Michaela K Back
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55128, Germany
| | - Jennifer Just
- TU Braunschweig, Zoological Institute, Braunschweig, 38106, Germany
| | - Lara Kilian
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Susanne Erdinger
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Marija Banicevic
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Lena Rehra
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Fadi Almouhanna
- Institute of Physiology and Pathophysiology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | - Martina Nigri
- Institute of Anatomy, University of Zurich, Zurich, 8057, Switzerland
| | - David P Wolfer
- Institute of Anatomy, University of Zurich, Zurich, 8057, Switzerland
- Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland
| | - Roman Spilger
- BioQuant Center, Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg and DKFZ, Heidelberg, 69120, Germany
| | - Karl Rohr
- BioQuant Center, Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg and DKFZ, Heidelberg, 69120, Germany
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| | | | - Jakob von Engelhardt
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, 55128, Germany
| | - Martin Korte
- TU Braunschweig, Zoological Institute, Braunschweig, 38106, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, 38124, Germany
| | - Ulrike C Müller
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht Karls Universität Heidelberg, Heidelberg, 69120, Germany
| |
Collapse
|
22
|
Gómez-Isla T, Frosch MP. Lesions without symptoms: understanding resilience to Alzheimer disease neuropathological changes. Nat Rev Neurol 2022; 18:323-332. [PMID: 35332316 PMCID: PMC10607925 DOI: 10.1038/s41582-022-00642-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
Abstract
Since the original description of amyloid-β plaques and tau tangles more than 100 years ago, these lesions have been considered the neuropathological hallmarks of Alzheimer disease (AD). The prevalence of plaques, tangles and dementia increases with age, and the lesions are considered to be causally related to the cognitive symptoms of AD. Current schemes for assessing AD lesion burden examine the distribution, abundance and characteristics of plaques and tangles at post mortem, yielding an estimate of the likelihood of cognitive impairment. Although this approach is highly predictive for most individuals, in some instances, a striking mismatch between lesions and symptoms can be observed. A small subset of individuals harbour a high burden of plaques and tangles at autopsy, which would be expected to have had devastating clinical consequences, but remain at their cognitive baseline, indicating 'resilience'. The study of these brains might provide the key to understanding the 'black box' between the accumulation of plaques and tangles and cognitive impairment, and show the way towards disease-modifying treatments for AD. In this Review, we begin by considering the heterogeneity of clinical manifestations associated with the presence of plaques and tangles, and then focus on insights derived from the rare yet informative individuals who display high amounts of amyloid and tau deposition in their brains (observed directly at autopsy) without manifesting dementia during life. The resilient response of these individuals to the gradual accumulation of plaques and tangles has potential implications for assessing an individual's risk of AD and for the development of interventions aimed at preserving cognition.
Collapse
Affiliation(s)
- Teresa Gómez-Isla
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA.
| | - Matthew P Frosch
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
23
|
Permanne B, Sand A, Ousson S, Nény M, Hantson J, Schubert R, Wiessner C, Quattropani A, Beher D. O-GlcNAcase Inhibitor ASN90 is a Multimodal Drug Candidate for Tau and α-Synuclein Proteinopathies. ACS Chem Neurosci 2022; 13:1296-1314. [PMID: 35357812 PMCID: PMC9026285 DOI: 10.1021/acschemneuro.2c00057] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Neurodegenerative proteinopathies are characterized by the intracellular formation of insoluble and toxic protein aggregates in the brain that are closely linked to disease progression. In Alzheimer's disease and in rare tauopathies, aggregation of the microtubule-associated tau protein leads to the formation of neurofibrillary tangles (NFT). In Parkinson's disease (PD) and other α-synucleinopathies, intracellular Lewy bodies containing aggregates of α-synuclein constitute the pathological hallmark. Inhibition of the glycoside hydrolase O-GlcNAcase (OGA) prevents the removal of O-linked N-acetyl-d-glucosamine (O-GlcNAc) moieties from intracellular proteins and has emerged as an attractive therapeutic approach to prevent the formation of tau pathology. Like tau, α-synuclein is known to be modified with O-GlcNAc moieties and in vitro these have been shown to prevent its aggregation and toxicity. Here, we report the preclinical discovery and development of a novel small molecule OGA inhibitor, ASN90. Consistent with the substantial exposure of the drug and demonstrating target engagement in the brain, the clinical OGA inhibitor ASN90 promoted the O-GlcNAcylation of tau and α-synuclein in brains of transgenic mice after daily oral dosing. Across human tauopathy mouse models, oral administration of ASN90 prevented the development of tau pathology (NFT formation), functional deficits in motor behavior and breathing, and increased survival. In addition, ASN90 slowed the progression of motor impairment and reduced astrogliosis in a frequently utilized α-synuclein-dependent preclinical rodent model of PD. These findings provide a strong rationale for the development of OGA inhibitors as disease-modifying agents in both tauopathies and α-synucleinopathies. Since tau and α-synuclein pathologies frequently co-exist in neurodegenerative diseases, OGA inhibitors represent unique, multimodal drug candidates for further clinical development.
Collapse
Affiliation(s)
- Bruno Permanne
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Astrid Sand
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Solenne Ousson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Maud Nény
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Jennifer Hantson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Ryan Schubert
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Christoph Wiessner
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Anna Quattropani
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Dirk Beher
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| |
Collapse
|
24
|
Pang K, Jiang R, Zhang W, Yang Z, Li LL, Shimozawa M, Tambaro S, Mayer J, Zhang B, Li M, Wang J, Liu H, Yang A, Chen X, Liu J, Winblad B, Han H, Jiang T, Wang W, Nilsson P, Guo W, Lu B. An App knock-in rat model for Alzheimer's disease exhibiting Aβ and tau pathologies, neuronal death and cognitive impairments. Cell Res 2022; 32:157-175. [PMID: 34789895 PMCID: PMC8807612 DOI: 10.1038/s41422-021-00582-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
A major obstacle in Alzheimer's disease (AD) research is the lack of predictive and translatable animal models that reflect disease progression and drug efficacy. Transgenic mice overexpressing amyloid precursor protein (App) gene manifest non-physiological and ectopic expression of APP and its fragments in the brain, which is not observed in AD patients. The App knock-in mice circumvented some of these problems, but they do not exhibit tau pathology and neuronal death. We have generated a rat model, with three familiar App mutations and humanized Aβ sequence knocked into the rat App gene. Without altering the levels of full-length APP and other APP fragments, this model exhibits pathologies and disease progression resembling those in human patients: deposit of Aβ plaques in relevant brain regions, microglia activation and gliosis, progressive synaptic degeneration and AD-relevant cognitive deficits. Interestingly, we have observed tau pathology, neuronal apoptosis and necroptosis and brain atrophy, phenotypes rarely seen in other APP models. This App knock-in rat model may serve as a useful tool for AD research, identifying new drug targets and biomarkers, and testing therapeutics.
Collapse
Affiliation(s)
- Keliang Pang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Richeng Jiang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Lin-Lin Li
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Mayer
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Baogui Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Man Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiesi Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ailing Yang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xi Chen
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiazheng Liu
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Hua Han
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Weiwen Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
25
|
Reactive astrocytes acquire neuroprotective as well as deleterious signatures in response to Tau and Aß pathology. Nat Commun 2022; 13:135. [PMID: 35013236 PMCID: PMC8748982 DOI: 10.1038/s41467-021-27702-w] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) alters astrocytes, but the effect of Aß and Tau pathology is poorly understood. TRAP-seq translatome analysis of astrocytes in APP/PS1 ß-amyloidopathy and MAPTP301S tauopathy mice revealed that only Aß influenced expression of AD risk genes, but both pathologies precociously induced age-dependent changes, and had distinct but overlapping signatures found in human post-mortem AD astrocytes. Both Aß and Tau pathology induced an astrocyte signature involving repression of bioenergetic and translation machinery, and induction of inflammation pathways plus protein degradation/proteostasis genes, the latter enriched in targets of inflammatory mediator Spi1 and stress-activated cytoprotective Nrf2. Astrocyte-specific Nrf2 expression induced a reactive phenotype which recapitulated elements of this proteostasis signature, reduced Aß deposition and phospho-tau accumulation in their respective models, and rescued brain-wide transcriptional deregulation, cellular pathology, neurodegeneration and behavioural/cognitive deficits. Thus, Aß and Tau induce overlapping astrocyte profiles associated with both deleterious and adaptive-protective signals, the latter of which can slow patho-progression.
Collapse
|
26
|
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
|
27
|
Puigdellívol M, Milde S, Vilalta A, Cockram TOJ, Allendorf DH, Lee JY, Dundee JM, Pampuščenko K, Borutaite V, Nuthall HN, Brelstaff JH, Spillantini MG, Brown GC. The microglial P2Y 6 receptor mediates neuronal loss and memory deficits in neurodegeneration. Cell Rep 2021; 37:110148. [PMID: 34965424 PMCID: PMC8733854 DOI: 10.1016/j.celrep.2021.110148] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/01/2021] [Accepted: 11/29/2021] [Indexed: 12/29/2022] Open
Abstract
Microglia are implicated in neurodegeneration, potentially by phagocytosing neurons, but it is unclear how to block the detrimental effects of microglia while preserving their beneficial roles. The microglial P2Y6 receptor (P2Y6R) - activated by extracellular UDP released by stressed neurons - is required for microglial phagocytosis of neurons. We show here that injection of amyloid beta (Aβ) into mouse brain induces microglial phagocytosis of neurons, followed by neuronal and memory loss, and this is all prevented by knockout of P2Y6R. In a chronic tau model of neurodegeneration (P301S TAU mice), P2Y6R knockout prevented TAU-induced neuronal and memory loss. In vitro, P2Y6R knockout blocked microglial phagocytosis of live but not dead targets and reduced tau-, Aβ-, and UDP-induced neuronal loss in glial-neuronal cultures. Thus, the P2Y6 receptor appears to mediate Aβ- and tau-induced neuronal and memory loss via microglial phagocytosis of neurons, suggesting that blocking this receptor may be beneficial in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK; Department of Biomedicine, School of Medicine, Institute of Neuroscience, University of Barcelona, 08036 Barcelona, Spain
| | - Stefan Milde
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Anna Vilalta
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Tom O J Cockram
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Jeffrey Y Lee
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Jacob M Dundee
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Katryna Pampuščenko
- Neuroscience Institute, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Hugh N Nuthall
- Neuroscience, Eli Lilly Research & Development, Windlesham, Surrey GU20 6PH, UK
| | - Jack H Brelstaff
- Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK.
| |
Collapse
|
28
|
Tau aggregation and its relation to selected forms of neuronal cell death. Essays Biochem 2021; 65:847-857. [PMID: 34897457 PMCID: PMC8709892 DOI: 10.1042/ebc20210030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
How neurons die in neurodegenerative diseases is still unknown. The distinction between apoptosis as a genetically controlled mechanism, and necrosis, which was viewed as an unregulated process, has blurred with the ever-increasing number of necrotic-like death subroutines underpinned by genetically defined pathways. It is therefore pertinent to ask whether any of them apply to neuronal cell death in tauopathies. Although Alzheimer's disease (AD) is the most prevalent tauopathy, tauopathies comprise an array of over 30 diseases in which the cytoplasmic protein tau aggregates in neurons, and also, in some diseases, in glia. Animal models have sought to distil the contribution of tau aggregation to the cell death process but despite intensive research, no one mechanism of cell death has been unequivocally defined. The process of tau aggregation, and the fibrillar structures that form, touch on so many cellular functions that there is unlikely to be a simple linear pathway of death; as one is blocked another is likely to take the lead. It is timely to ask how far we have advanced into defining whether any of the molecular players in the new death subroutines participate in the death process. Here we briefly review the currently known cell death routines and explore what is known about their participation in tau aggregation-related cell death. We highlight the involvement of cell autonomous and the more recent non-cell autonomous pathways that may enhance tau-aggregate toxicity, and discuss recent findings that implicate microglial phagocytosis of live neurons with tau aggregates as a mechanism of death.
Collapse
|
29
|
Mirzaei N, Davis N, Chau TW, Sastre M. Astrocyte Reactivity in Alzheimer's Disease: Therapeutic Opportunities to Promote Repair. Curr Alzheimer Res 2021; 19:1-15. [PMID: 34719372 DOI: 10.2174/1567205018666211029164106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/02/2021] [Accepted: 07/31/2021] [Indexed: 11/22/2022]
Abstract
Astrocytes are fast climbing the ladder of importance in neurodegenerative disorders, particularly in Alzheimer's disease (AD), with the prominent presence of reactive astrocytes sur- rounding amyloid β- plaques, together with activated microglia. Reactive astrogliosis, implying morphological and molecular transformations in astrocytes, seems to precede neurodegeneration, suggesting a role in the development of the disease. Single-cell transcriptomics has recently demon- strated that astrocytes from AD brains are different from "normal" healthy astrocytes, showing dys- regulations in areas such as neurotransmitter recycling, including glutamate and GABA, and im- paired homeostatic functions. However, recent data suggest that the ablation of astrocytes in mouse models of amyloidosis results in an increase in amyloid pathology as well as in the inflammatory profile and reduced synaptic density, indicating that astrocytes mediate neuroprotective effects. The idea that interventions targeting astrocytes may have great potential for AD has therefore emerged, supported by a range of drugs and stem cell transplantation studies that have successfully shown a therapeutic effect in mouse models of AD. In this article, we review the latest reports on the role and profile of astrocytes in AD brains and how manipulation of astrocytes in animal mod- els has paved the way for the use of treatments enhancing astrocytic function as future therapeutic avenues for AD.
Collapse
Affiliation(s)
- Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048. United States
| | - Nicola Davis
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Tsz Wing Chau
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammer-smith Hospital, Du Cane Road, LondonW12 0NN. United Kingdom
| |
Collapse
|
30
|
Do Carmo S, Spillantini MG, Cuello AC. Editorial: Tau Pathology in Neurological Disorders. Front Neurol 2021; 12:754669. [PMID: 34630315 PMCID: PMC8497747 DOI: 10.3389/fneur.2021.754669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/26/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, United Kingdom
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Pharmacology, Oxford University, Oxford, United Kingdom
| |
Collapse
|
31
|
Hartnell IJ, Blum D, Nicoll JAR, Dorothee G, Boche D. Glial cells and adaptive immunity in frontotemporal dementia with tau pathology. Brain 2021; 144:724-745. [PMID: 33527991 DOI: 10.1093/brain/awaa457] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is involved in the aetiology of many neurodegenerative disorders including Alzheimer's disease, Parkinson's disease and motor neuron disease. Whether neuroinflammation also plays an important role in the pathophysiology of frontotemporal dementia is less well known. Frontotemporal dementia is a heterogeneous classification that covers many subtypes, with the main pathology known as frontotemporal lobar degeneration. The disease can be categorized with respect to the identity of the protein that causes the frontotemporal lobar degeneration in the brain. The most common subgroup describes diseases caused by frontotemporal lobar degeneration associated with tau aggregation, also known as primary tauopathies. Evidence suggests that neuroinflammation may play a role in primary tauopathies with genome-wide association studies finding enrichment of genetic variants associated with specific inflammation-related gene loci. These loci are related to both the innate immune system, including brain resident microglia, and the adaptive immune system through possible peripheral T-cell involvement. This review discusses the genetic evidence and relates it to findings in animal models expressing pathogenic tau as well as to post-mortem and PET studies in human disease. Across experimental paradigms, there seems to be a consensus regarding the involvement of innate immunity in primary tauopathies, with increased microglia and astrocyte density and/or activation, as well as increases in pro-inflammatory markers. Whilst it is less clear as to whether inflammation precedes tau aggregation or vice versa; there is strong evidence to support a microglial contribution to the propagation of hyperphosphorylated in tau frontotemporal lobar degeneration associated with tau aggregation. Experimental evidence-albeit limited-also corroborates genetic data pointing to the involvement of cellular adaptive immunity in primary tauopathies. However, it is still unclear whether brain recruitment of peripheral immune cells is an aberrant result of pathological changes or a physiological aspect of the neuroinflammatory response to the tau pathology.
Collapse
Affiliation(s)
- Iain J Hartnell
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - David Blum
- University of Lille, Inserm, CHU-Lille, UMR-S 1172-Lille Neuroscience and Cognition, Lille, France.,Alzheimer & Tauopathies, LabEx DISTALZ, France
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Guillaume Dorothee
- Inserm, Sorbonne University, UMRS 938 Saint-Antoine Research Center, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
32
|
Ungerleider K, Beck J, Lissa D, Turnquist C, Horikawa I, Harris BT, Harris CC. Astrocyte senescence and SASP in neurodegeneration: tau joins the loop. Cell Cycle 2021; 20:752-764. [PMID: 33818291 DOI: 10.1080/15384101.2021.1909260] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tau accumulation is a core component of Alzheimer's disease and other neurodegenerative tauopathies. While tau's impact on neurons is a major area of research, the effect of extracellular tau on astrocytes is largely unknown. This article summarizes our recent studies showing that astrocyte senescence plays a critical role in neurodegenerative diseases and integrates extracellular tau into the regulatory loop of senescent astrocyte-mediated neurotoxicity. Human astrocytes in vitro undergoing senescence were shown to acquire the inflammatory senescence-associated secretory phenotype (SASP) and toxicity to neurons, which may recapitulate aging- and disease-associated neurodegeneration. Here, we show that human astrocytes exposed to extracellular tau in vitro also undergo cellular senescence and acquire a neurotoxic SASP (e.g. IL-6 secretion), with oxidative stress response (indicated by upregulated NRF2 target genes) and a possible activation of inflammasome (indicated by upregulated ASC and IL-1β). These findings suggest that senescent astrocytes induced by various conditions and insults, including tau exposure, may represent a therapeutic target to inhibit or delay the progression of neurodegenerative diseases. We also discuss the pathological activity of extracellular tau in microglia and astrocytes, the disease relevance and diversity of tau forms, therapeutics targeting senescence in neurodegeneration, and the roles of p53 and its isoforms in astrocyte-mediated neurotoxicity and neuroprotection.
Collapse
Affiliation(s)
- Kyra Ungerleider
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Comparative Pathobiology, Purdue University, West Layfette, Indiana, USA
| | - Delphine Lissa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Casmir Turnquist
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,University of Oxford Medical School, John Radcliffe Hospital, Oxford, UK
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brent T Harris
- Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Sidoryk-Wegrzynowicz M, Strużyńska L. Dysfunctional glia: contributors to neurodegenerative disorders. Neural Regen Res 2021; 16:218-222. [PMID: 32859767 PMCID: PMC7896233 DOI: 10.4103/1673-5374.290877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Astrocytes are integral components of the central nervous system, where they are involved in numerous functions critical for neuronal development and functioning, including maintenance of blood-brain barrier, formation of synapses, supporting neurons with nutrients and trophic factors, and protecting them from injury. These roles are markedly affected in the course of chronic neurodegenerative disorders, often before the onset of the disease. In this review, we summarize the recent findings supporting the hypothesis that astrocytes play a fundamental role in the processes contributing to neurodegeneration. We focus on α-synucleinopathies and tauopathies as the most common neurodegenerative diseases. The mechanisms implicated in the development and progression of these disorders appear not to be exclusively neuronal, but are often related to the astrocytic-neuronal integrity and the response of astrocytes to the altered microglial function. A profound understanding of the multifaceted functions of astrocytes and identification of their communication pathways with neurons and microglia in health and in the disease is of critical significance for the development of novel mechanism-based therapies against neurodegenerative disorders.
Collapse
Affiliation(s)
- Marta Sidoryk-Wegrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
34
|
Spanos F, Liddelow SA. An Overview of Astrocyte Responses in Genetically Induced Alzheimer's Disease Mouse Models. Cells 2020; 9:E2415. [PMID: 33158189 PMCID: PMC7694249 DOI: 10.3390/cells9112415] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite many years of intense research, there is currently still no effective treatment. Multiple cell types contribute to disease pathogenesis, with an increasing body of data pointing to the active participation of astrocytes. Astrocytes play a pivotal role in the physiology and metabolic functions of neurons and other cells in the central nervous system. Because of their interactions with other cell types, astrocyte functions must be understood in their biologic context, thus many studies have used mouse models, of which there are over 190 available for AD research. However, none appear able to fully recapitulate the many functional changes in astrocytes reported in human AD brains. Our review summarizes the observations of astrocyte biology noted in mouse models of familial and sporadic AD. The limitations of AD mouse models will be discussed and current attempts to overcome these disadvantages will be described. With increasing understanding of the non-neuronal contributions to disease, the development of new methods and models will provide further insights and address important questions regarding the roles of astrocytes and other non-neuronal cells in AD pathophysiology. The next decade will prove to be full of exciting opportunities to address this devastating disease.
Collapse
Affiliation(s)
- Fokion Spanos
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
35
|
Weninger S, Sperling B, Alexander R, Ivarsson M, Menzies FM, Powchik P, Weber CJ, Altar CA, Crystal RG, Haggarty SJ, Loring J, Bain LJ, Carrillo MC. Active immunotherapy and alternative therapeutic modalities for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2020; 6:e12090. [PMID: 33083513 PMCID: PMC7550557 DOI: 10.1002/trc2.12090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022]
Abstract
As knowledge of Alzheimer's disease (AD) progression improves, the field has recognized the need to diversify the pipeline, broaden strategies and approaches to therapies, as well as delivery mechanisms. A better understanding of the earliest biological processes of AD/dementia would help inform drug target selection. Currently there are a number of programs exploring these alternate avenues. This meeting will allow experts in the field (academia, industry, government) to provide perspectives and experiences that can help elucidate what the pipeline looks like today and what avenues hold promise in developing new therapies across the stages of AD. The focus here is on Active Immunotherapies and Alternative Therapeutic Modalities. This topic includes active vaccines, antisense oligomers, and cell-based therapy among others, and highlights new clinical developments that utilize these modalities.
Collapse
Affiliation(s)
| | | | - Robert Alexander
- Takeda Pharmaceuticals International Co. Cambridge Massachusetts USA
| | - Magnus Ivarsson
- Rodin Therapeutics 300 Technology Square Cambridge Massachusetts USA
| | | | - Peter Powchik
- United Neuroscience 9 Exchange Place, I. F. S. C Dublin Ireland
| | | | | | - Ronald G Crystal
- Department of Genetic Medicine Weill Cornell Medicine New York New York USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory Center for Genomic Medicine Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| | | | - Lisa J Bain
- Independent Science Writer Elverson Pennsylvania USA
| | | |
Collapse
|
36
|
Astroglial contribution to tau-dependent neurodegeneration. Biochem J 2020; 476:3493-3504. [PMID: 31774919 DOI: 10.1042/bcj20190506] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 01/31/2023]
Abstract
Astrocytes, by maintaining an optimal environment for neuronal function, play a critical role in proper function of mammalian nervous system. They regulate synaptic transmission and plasticity and protect neurons against toxic insults. Astrocytes and neurons interact actively via glutamine-glutamate cycle (GGC) that supports neuronal metabolic demands and neurotransmission. GGC deficiency may be involved in different diseases of the brain, where impaired astrocytic control of glutamate homeostasis contributes to neuronal dysfunction. This includes tau-dependent neurodegeneration, where astrocytes lose key molecules involved in regulation of glutamate/glutamine homeostasis, neuronal survival and synaptogenesis. Astrocytic dysfunction in tauopathy appears to precede neurodegeneration and overt tau neuropathology such as phosphorylation, aggregation and formation of neurofibrillary tangles. In this review, we summarize recent studies demonstrating that activation of astrocytes is strictly associated with neurodegenerative processes including those involved in tau related pathology. We propose that astrocytic dysfunction, by disrupting the proper neuron-glia signalling early in the disease, significantly contributes to tauopathy pathogenesis.
Collapse
|
37
|
Hampton DW, Amor S, Story D, Torvell M, Bsibsi M, van Noort JM, Chandran S. HspB5 Activates a Neuroprotective Glial Cell Response in Experimental Tauopathy. Front Neurosci 2020; 14:574. [PMID: 32595446 PMCID: PMC7300208 DOI: 10.3389/fnins.2020.00574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/11/2020] [Indexed: 12/27/2022] Open
Abstract
Progressive neuronal death during tauopathies is associated with aggregation of modified, truncated or mutant forms of tau protein. Such aggregates are neurotoxic, promote spreading of tau aggregation, and trigger release of pro-inflammatory factors by glial cells. Counteracting such pathogenic effects of tau by simultaneously inhibiting protein aggregation as well as pro-inflammatory glial cell responses would be of significant therapeutic interest. Here, we examined the use of the small heat-shock protein HspB5 for this purpose. As a molecular chaperone, HspB5 counteracts aggregation of a wide range of abnormal proteins. As a TLR2 agonist, it selectively activates protective responses by CD14-expressing myeloid cells including microglia. We show that intracerebral infusion of HspB5 in transgenic mice with selective neuronal expression of mutant human P301S tau has significant neuroprotective effects in the superficial, frontal cortical layers. Underlying these effects at least in part, HspB5 induces several potent neuroprotective mediators in both astrocytes and microglia including neurotrophic factors and increased potential for removal of glutamate. Together, these findings highlight the potentially broad therapeutic potential of HspB5 in neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- David W Hampton
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Sandra Amor
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | - David Story
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Megan Torvell
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | | | | | - Siddarthan Chandran
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,UK Dementia Research Institute, Edinburgh University, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Teravskis PJ, Ashe KH, Liao D. The Accumulation of Tau in Postsynaptic Structures: A Common Feature in Multiple Neurodegenerative Diseases? Neuroscientist 2020; 26:503-520. [PMID: 32389059 DOI: 10.1177/1073858420916696] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increasingly, research suggests that neurodegenerative diseases and dementias are caused not by unique, solitary cellular mechanisms, but by multiple contributory mechanisms manifesting as heterogeneous clinical presentations. However, diverse neurodegenerative diseases also share common pathological hallmarks and cellular mechanisms. One such mechanism involves the redistribution of the microtubule associated protein tau from the axon into the somatodendritic compartment of neurons, followed by the mislocalization of tau into dendritic spines, resulting in postsynaptic functional deficits. Here we review various signaling pathways that trigger the redistribution of tau to the cell body and dendritic tree, and its mislocalization to dendritic spines. The convergence of multiple pathways in different disease models onto this final common pathway suggests that it may be an attractive pathway to target for developing new treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter J Teravskis
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.,University of Minnesota Medical School, Minneapolis, MN, USA
| | - Karen H Ashe
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Budd Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA.,Geriatric Research Education and Clinical Center, Veterans Affairs Medical Center, Minneapolis, MN, USA
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
39
|
Abstract
Astrocytes contribute to the pathogenesis of neurodegenerative proteinopathies as influencing neuronal degeneration or neuroprotection, and also act as potential mediators of the propagation or elimination of disease-associated proteins. Protein astrogliopathies can be observed in different forms of neurodegenerative conditions. Morphological characterization of astrogliopathy is used only for the classification of tauopathies. Currently, at least six types of astrocytic tau pathologies are distinguished. Astrocytic plaques (AP), tufted astrocytes (TAs), ramified astrocytes (RA), and globular astroglial inclusions are seen predominantly in primary tauopathies, while thorn-shaped astrocytes (TSA) and granular/fuzzy astrocytes (GFA) are evaluated in aging-related tau astrogliopathy (ARTAG). ARTAG can be seen in the white and gray matter and subpial, subependymal, and perivascular locations. Some of these overlap with the features of tau pathology seen in Chronic traumatic encephalopathy (CTE). Furthermore, gray matter ARTAG shares features with primary tauopathy-related astrocytic tau pathology. Sequential distribution patterns have been described for tau astrogliopathies. Importantly, astrocytic tau pathology in primary tauopathies can be observed in brain areas without neuronal tau deposition. The various morphologies of tau astrogliopathy might reflect a role in the propagation of pathological tau protein, an early response to a yet unidentified neurodegeneration-inducing event, or, particularly for ARTAG, a response to a repeated or prolonged pathogenic process such as blood-brain barrier dysfunction or local mechanical impact. The concept of tau astrogliopathies and ARTAG facilitated communication among research disciplines and triggered the investigation of the significance of astrocytic lesions in neurodegenerative conditions.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
| |
Collapse
|
40
|
Abstract
The symptoms of Alzheimer disease reflect a loss of neural circuit integrity in the brain, but neurons do not work in isolation. Emerging evidence suggests that the intricate balance of interactions between neurons, astrocytes, microglia and vascular cells required for healthy brain function becomes perturbed during the disease, with early changes likely protecting neural circuits from damage, followed later by harmful effects when the balance cannot be restored. Moving beyond a neuronal focus to understand the complex cellular interactions in Alzheimer disease and how these change throughout the course of the disease may provide important insight into developing effective therapeutics.
Collapse
|
41
|
Boese AC, Hamblin MH, Lee JP. Neural stem cell therapy for neurovascular injury in Alzheimer's disease. Exp Neurol 2019; 324:113112. [PMID: 31730762 DOI: 10.1016/j.expneurol.2019.113112] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by progressive neurodegeneration leading to severe cognitive decline and eventual death. AD pathophysiology is complex, but neurotoxic accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau are believed to be main drivers of neurodegeneration in AD. The formation and deposition of Aβ plaques occurs in the brain parenchyma as well as in the cerebral vasculature. Thus, proper blood-brain barrier (BBB) and cerebrovascular functioning are crucial for clearance of Aβ from the brain, and neurovascular dysfunction may be a critical component of AD development. Further, neuroinflammation and dysfunction of angiogenesis, neurogenesis, and neurorestorative capabilities play a role in AD pathophysiology. Currently, there is no effective treatment to prevent or restore loss of brain tissue and cognitive decline in patients with AD. Based on multifactorial and complex pathophysiological cascades in multiple Alzheimer's disease stages, effective AD therapies need to focus on targeting early AD pathology and preserving cerebrovascular function. Neural stem cells (NSCs) participate extensively in mammalian brain homeostasis and repair and exhibit pleiotropic intrinsic properties that likely make them attractive candidates for the treatment of AD. In the review, we summarize the current advances in knowledge regarding neurovascular aspects of AD-related neurodegeneration and discuss multiple actions of NSCs from preclinical studies of AD to evaluate their potential for future clinical treatment of AD.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
42
|
Torvell M, Hampton DW, Connick P, MacLullich AMJ, Cunningham C, Chandran S. A single systemic inflammatory insult causes acute motor deficits and accelerates disease progression in a mouse model of human tauopathy. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:579-591. [PMID: 31650014 PMCID: PMC6804509 DOI: 10.1016/j.trci.2019.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Introduction Neuroinflammation, which contributes to neurodegeneration, is a consistent hallmark of dementia. Emerging evidence suggests that systemic inflammation also contributes to disease progression. Methods The ability of systemically administered lipopolysaccharide (LPS - 500 μg/kg) to effect acute and chronic behavioural changes in C57BL/6 and P301S tauopathy mice was assessed. Markers of pathology were assessed in the brain and spinal cord. Results P301S mice display regional microgliosis. Systemic LPS treatment induced exaggerated acute sickness behaviour and motor dysfunction in P301S mice compared with wild-type controls and advanced the onset and accelerated chronic decline. LPS treatment was associated with increased tau pathology 24 hours after LPS injection and spinal cord microgliosis at the end stage. Discussion This is the first demonstration that a single systemic inflammatory episode causes exaggerated acute functional impairments and accelerates the long-term trajectory of functional decline associated with neurodegeneration in a mouse model of human tauopathy. The findings have relevance to management of human dementias. P301S microgliosis is regional; activation occurs in the spinal cord but not in the cortex. Systemic LPS injection caused acute neurological deficits in P301S mice. This was associated with increased tau pathology 24 hours after LPS injection. This was independent of microglial priming as measured by IL-1β hyperexpression. LPS injection advanced the onset of chronic decline in P301S mice.
Collapse
Affiliation(s)
- Megan Torvell
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Cardiff, Cardiff, UK
| | - David W Hampton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK
| | - Peter Connick
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian, UK
| | - Alasdair M J MacLullich
- Edinburgh Delirium Research Group, Geriatric Medicine, University of Edinburgh, Edinburgh, UK
| | - Colm Cunningham
- Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK.,The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, Midlothian, UK
| |
Collapse
|
43
|
Mancuso R, Fryatt G, Cleal M, Obst J, Pipi E, Monzón-Sandoval J, Ribe E, Winchester L, Webber C, Nevado A, Jacobs T, Austin N, Theunis C, Grauwen K, Daniela Ruiz E, Mudher A, Vicente-Rodriguez M, Parker CA, Simmons C, Cash D, Richardson J. CSF1R inhibitor JNJ-40346527 attenuates microglial proliferation and neurodegeneration in P301S mice. Brain 2019; 142:3243-3264. [PMID: 31504240 PMCID: PMC6794948 DOI: 10.1093/brain/awz241] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 11/12/2022] Open
Abstract
Neuroinflammation and microglial activation are significant processes in Alzheimer's disease pathology. Recent genome-wide association studies have highlighted multiple immune-related genes in association with Alzheimer's disease, and experimental data have demonstrated microglial proliferation as a significant component of the neuropathology. In this study, we tested the efficacy of the selective CSF1R inhibitor JNJ-40346527 (JNJ-527) in the P301S mouse tauopathy model. We first demonstrated the anti-proliferative effects of JNJ-527 on microglia in the ME7 prion model, and its impact on the inflammatory profile, and provided potential CNS biomarkers for clinical investigation with the compound, including pharmacokinetic/pharmacodynamics and efficacy assessment by TSPO autoradiography and CSF proteomics. Then, we showed for the first time that blockade of microglial proliferation and modification of microglial phenotype leads to an attenuation of tau-induced neurodegeneration and results in functional improvement in P301S mice. Overall, this work strongly supports the potential for inhibition of CSF1R as a target for the treatment of Alzheimer's disease and other tau-mediated neurodegenerative diseases.
Collapse
Affiliation(s)
- Renzo Mancuso
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Gemma Fryatt
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Madeleine Cleal
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Juliane Obst
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Elena Pipi
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Jimena Monzón-Sandoval
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Elena Ribe
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Laura Winchester
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Caleb Webber
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Alejo Nevado
- Department of Physiology Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford OX1 3PT, UK
| | - Tom Jacobs
- Janssen Research and Development, Turnhoutseweg 30, box 270, 2340 Beerse 1, Belgium
| | - Nigel Austin
- Janssen Research and Development, Turnhoutseweg 30, box 270, 2340 Beerse 1, Belgium
| | - Clara Theunis
- Janssen Neuroscience Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Karolien Grauwen
- Janssen Neuroscience Research and Development, Janssen Pharmaceutical Companies of Johnson and Johnson, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Eva Daniela Ruiz
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Amrit Mudher
- Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Marta Vicente-Rodriguez
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Christine A Parker
- Experimental Medicine Imaging, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Camilla Simmons
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Jill Richardson
- Neurosciences Therapeutic Area, GlaxoSmithKline R&D, Stevenage, UK
| |
Collapse
|
44
|
Dunn-Meynell AA, Dowling P, Marchese M, Rodriguez E, Blumberg B, Choi YB, Gaindh D, Lu W. In vivo Bioluminescence Imaging Used to Monitor Disease Activity and Therapeutic Response in a Mouse Model of Tauopathy. Front Aging Neurosci 2019; 11:252. [PMID: 31572168 PMCID: PMC6751306 DOI: 10.3389/fnagi.2019.00252] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/26/2019] [Indexed: 01/03/2023] Open
Abstract
Many studies of tauopathy use transgenic mice that overexpress the P301S mutant form of tau. Neuronal damage in these mice is associated with astrogliosis and induction of glial fibrillary acidic protein (GFAP) expression. GFAP-luc transgenic mice express firefly luciferase under the GFAP promoter, allowing bioluminescence to be measured non-invasively as a surrogate biomarker for astrogliosis. We bred double transgenic mice possessing both P301S and GFAP-luc cassettes and compared them to control mice bearing only the GFAP-luc transgene. We used serial bioluminescent images to define the onset and the time course of astrogliosis in these mice and this was correlated with the development of clinical deficit. Mice containing both GFAP-luc and P301S transgenes showed increased luminescence indicative of astroglial activation in the brain and spinal cord. Starting at 5 months old, the onset of clinical deterioration in these mice corresponded closely to the initial rise in the luminescent signal. Post mortem analysis showed the elevated luminescence was correlated with hyperphosphorylated tau deposition in the hippocampus of double transgenic mice. We used this method to determine the therapeutic effect of JM4 peptide [a small peptide immunomodulatory agent derived from human erythropoietin (EPO)] on double transgenic mice. JM4 treatment significantly decreased the intensity of luminescence, neurological deficit and hyperphosphorylated tau in mice with both the P301S and GFAP-luc transgenes. These findings indicate that bioluminescence imaging (BLI) is a powerful tool for quantifying GFAP expression in living P301S mice and can be used as a noninvasive biomarker of tau-induced neurodegeneration in preclinical therapeutic trials.
Collapse
Affiliation(s)
- Ambrose A. Dunn-Meynell
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Peter Dowling
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Michelle Marchese
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Esther Rodriguez
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Benjamin Blumberg
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| | - Yun-Beom Choi
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Deeya Gaindh
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
- Department of Neurology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, United States
| | - Wei Lu
- Neurology Service, VA New Jersey Health Care System, East Orange, NJ, United States
| |
Collapse
|
45
|
Knock-in of Mutated hTAU Causes Insulin Resistance, Inflammation and Proteostasis Disturbance in a Mouse Model of Frontotemporal Dementia. Mol Neurobiol 2019; 57:539-550. [PMID: 31396860 PMCID: PMC6968995 DOI: 10.1007/s12035-019-01722-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/26/2019] [Indexed: 12/21/2022]
Abstract
Diabetes and obesity have been implicated as risk factors for dementia. However, metabolic mechanisms and associated signalling pathways have not been investigated in detail in frontotemporal dementia. We therefore here characterised physiological, behavioural and molecular phenotypes of 3- and 8-month-old male tau knock-in (PLB2TAU) vs wild-type (PLBWT) mice. Homecage analysis suggested intact habituation but a dramatic reduction in exploratory activity in PLB2TAU mice. Deficits in motor strength were also observed. At 3 months, PLB2TAU mice displayed normal glucose handling but developed hyperglycaemia at 8 months, suggesting a progressive diabetic phenotype. Brain, liver and muscle tissue analyses confirmed tissue-specific deregulation of metabolic and homeostatic pathways. In brain, increased levels of phosphorylated tau and inflammation were detected alongside reduced ER regulatory markers, overall suggesting a downregulation in essential cellular defence pathways. We suggest that subtle neuronal expression of mutated human tau is sufficient to disturb systems metabolism and protein handling. Whether respective dysfunctions in tauopathy patients are also a consequence of tau pathology remains to be confirmed, but could offer new avenues for therapeutic interventions.
Collapse
|
46
|
Chakari-Khiavi F, Dolati S, Chakari-Khiavi A, Abbaszadeh H, Aghebati-Maleki L, Pourlak T, Mehdizadeh A, Yousefi M. Prospects for the application of mesenchymal stem cells in Alzheimer's disease treatment. Life Sci 2019; 231:116564. [PMID: 31202840 DOI: 10.1016/j.lfs.2019.116564] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) as a dementia and neurodegenerative disease, is mostly prevalent among people more than 65 years. AD is mostly manifested in the form of degraded mental function, such as losing memory and impaired cognitive function. Due to inefficiency of traditional pharmacological therapeutic approaches with no long-term cure, cell therapy can be considered as a capable approach in AD management. Therapies based on mesenchymal stem cells (MSCs) have provided hopeful results in experimental models regarding several disorders. MSCs enhance the levels of functional recoveries in pathologic experimental models of central nervous system (CNS) and are being investigated in clinical trials in neurological disorders. However, there is limited knowledge on the protective capabilities of MSCs in AD management. Almost, several experiments have suggested positive effects of MSCs and helped to better understand of AD-related dementia mechanism. MSCs have the potential to be used in AD treatment through amyloid-β peptide (AB), Tau protein and cholinergic system. This review aimed to clarify the promising perspective of MSCs in the context of AD.
Collapse
Affiliation(s)
- Forough Chakari-Khiavi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Aref Chakari-Khiavi
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Hossein Abbaszadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Tannaz Pourlak
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran..
| |
Collapse
|
47
|
Sadick JS, Liddelow SA. Don't forget astrocytes when targeting Alzheimer's disease. Br J Pharmacol 2019; 176:3585-3598. [PMID: 30636042 DOI: 10.1111/bph.14568] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are essential for CNS health, regulating homeostasis, metabolism, and synaptic transmission. In addition to these and many other physiological roles, the pathological impact of astrocytes ("reactive astrocytes") in acute trauma and chronic disease like Alzheimer's disease (AD) is well established. Growing evidence supports a fundamental and active role of astrocytes in multiple neurodegenerative diseases. With a growing interest in normal astrocyte biology, and countless studies on changes in astrocyte function in the context of disease, it may be a surprise that no therapies exist incorporating astrocytes as key targets. Here, we examine unintentional effects of current AD therapies on astrocyte function and theorize how astrocytes may be intentionally targeted for more efficacious therapeutic outcomes. Given their integral role in normal neuronal functioning, incorporating astrocytes as key criteria for AD drug development can only lead to more effective therapies for the millions of AD sufferers worldwide. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Jessica S Sadick
- Neuroscience Institute, NYU Langone Medical Center, New York, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Langone Medical Center, New York, USA.,Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, USA.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Keaney J, Gasser J, Gillet G, Scholz D, Kadiu I. Inhibition of Bruton's Tyrosine Kinase Modulates Microglial Phagocytosis: Therapeutic Implications for Alzheimer's Disease. J Neuroimmune Pharmacol 2019; 14:448-461. [PMID: 30758770 PMCID: PMC6707957 DOI: 10.1007/s11481-019-09839-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
Bruton’s tyrosine kinase (BTK), a critical component of B cell receptor signaling, has recently been implicated in regulation of the peripheral innate immune response. However, the role of BTK in microglia, the resident innate immune cells of the central nervous system, and its involvement in the pathobiology of neurodegenerative disease has not been explored. Here we found that BTK is a key regulator of microglial phagocytosis. Using potent BTK inhibitors and small interfering RNA (siRNA) against BTK, we observed that blockade of BTK activity decreased activation of phospholipase gamma 2, a recently identified genetic risk factor in Alzheimer’s disease (AD), and reduced phagocytosis in rodent microglia and human monocyte-derived macrophages. Inhibition of BTK signaling also decreased microglial uptake of synaptosomes but did not have major impacts on other key microglial functions such as migration and cytokine release. Similarly, blocking BTK function ex vivo in acute brain slices reduced microglial phagocytosis and maintained numbers of resting microglia. In brain tissues from the 5xFAD mouse model of AD, levels of microglial BTK were elevated while in two gene expression datasets of post-mortem AD patient brain tissues, upregulation of BTK transcript was observed. Our study provides novel insights into the role of BTK in regulating microglial phagocytosis and uptake of synaptic structures and suggests that inhibiting microglial BTK may improve cognition in AD by preventing microglial activation and synaptic loss. Microglial-mediated synapse loss has been implicated in AD pathogenesis. Inhibition of BTK decreases activation of PLCγ2, a genetic risk factor in AD, and reduces microglial phagocytosis and uptake of synaptic structures. As such BTK inhibition may represent a therapeutic route to prevent microglial activation and synapse loss in AD ![]()
Collapse
Affiliation(s)
- James Keaney
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium.
| | - Julien Gasser
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium
| | - Gaëlle Gillet
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium
| | - Diana Scholz
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium
| | - Irena Kadiu
- Neuroscience Therapeutic Area, New Medicines, UCB Biopharma SPRL, Chemin du Foriest, 1420, Braine-l'Alleud, Belgium.
| |
Collapse
|
49
|
Jones MK, Lu B, Chen DZ, Spivia WR, Mercado AT, Ljubimov AV, Svendsen CN, Van Eyk JE, Wang S. In Vitro and In Vivo Proteomic Comparison of Human Neural Progenitor Cell-Induced Photoreceptor Survival. Proteomics 2019; 19:e1800213. [PMID: 30515959 PMCID: PMC6422354 DOI: 10.1002/pmic.201800213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/01/2018] [Indexed: 12/31/2022]
Abstract
Retinal degenerative diseases lead to blindness with few treatments. Various cell-based therapies are aimed to slow the progression of vision loss by preserving light-sensing photoreceptor cells. A subretinal injection of human neural progenitor cells (hNPCs) into the Royal College of Surgeons (RCS) rat model of retinal degeneration has aided in photoreceptor survival, though the mechanisms are mainly unknown. Identifying the retinal proteomic changes that occur following hNPC treatment leads to better understanding of neuroprotection. To mimic the retinal environment following hNPC injection, a co-culture system of retinas and hNPCs is developed. Less cell death occurs in RCS retinal tissue co-cultured with hNPCs than in retinas cultured alone, suggesting that hNPCs provide retinal protection in vitro. Comparison of ex vivo and in vivo retinas identifies nuclear factor (erythroid-derived 2)-like 2 (NRF2) mediated oxidative response signaling as an hNPC-induced pathway. This is the first study to compare proteomic changes following treatment with hNPCs in both an ex vivo and in vivo environment, further allowing the use of ex vivo modeling for mechanisms of retinal preservation. Elucidation of the protein changes in the retina following hNPC treatment may lead to the discovery of mechanisms of photoreceptor survival and its therapeutic for clinical applications.
Collapse
Affiliation(s)
- Melissa K. Jones
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Bin Lu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Dawn Z. Chen
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Weston R. Spivia
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
| | - Augustus T. Mercado
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Alexander V. Ljubimov
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Clive N. Svendsen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| | - Shaomei Wang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles
| |
Collapse
|
50
|
Beyer F, Samper Agrelo I, Küry P. Do Neural Stem Cells Have a Choice? Heterogenic Outcome of Cell Fate Acquisition in Different Injury Models. Int J Mol Sci 2019; 20:ijms20020455. [PMID: 30669690 PMCID: PMC6359747 DOI: 10.3390/ijms20020455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/14/2019] [Accepted: 01/18/2019] [Indexed: 12/19/2022] Open
Abstract
The adult mammalian central nervous system (CNS) is generally considered as repair restricted organ with limited capacities to regenerate lost cells and to successfully integrate them into damaged nerve tracts. Despite the presence of endogenous immature cell types that can be activated upon injury or in disease cell replacement generally remains insufficient, undirected, or lost cell types are not properly generated. This limitation also accounts for the myelin repair capacity that still constitutes the default regenerative activity at least in inflammatory demyelinating conditions. Ever since the discovery of endogenous neural stem cells (NSCs) residing within specific niches of the adult brain, as well as the description of procedures to either isolate and propagate or artificially induce NSCs from various origins ex vivo, the field has been rejuvenated. Various sources of NSCs have been investigated and applied in current neuropathological paradigms aiming at the replacement of lost cells and the restoration of functionality based on successful integration. Whereas directing and supporting stem cells residing in brain niches constitutes one possible approach many investigations addressed their potential upon transplantation. Given the heterogeneity of these studies related to the nature of grafted cells, the local CNS environment, and applied implantation procedures we here set out to review and compare their applied protocols in order to evaluate rate-limiting parameters. Based on our compilation, we conclude that in healthy CNS tissue region specific cues dominate cell fate decisions. However, although increasing evidence points to the capacity of transplanted NSCs to reflect the regenerative need of an injury environment, a still heterogenic picture emerges when analyzing transplantation outcomes in injury or disease models. These are likely due to methodological differences despite preserved injury environments. Based on this meta-analysis, we suggest future NSC transplantation experiments to be conducted in a more comparable way to previous studies and that subsequent analyses must emphasize regional heterogeneity such as accounting for differences in gray versus white matter.
Collapse
Affiliation(s)
- Felix Beyer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| | - Iria Samper Agrelo
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| |
Collapse
|