1
|
Solem MA, Pelzel R, Rozema NB, Brown TG, Reid E, Mansky RH, Gomez-Pastor R. Enhanced Hippocampal Spare Capacity in Q175DN Mice Despite Elevated mHTT Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618355. [PMID: 39464002 PMCID: PMC11507687 DOI: 10.1101/2024.10.14.618355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background Huntington's disease (HD) is a neurodegenerative disease resulting in devastating motor, cognitive, and psychiatric deficits. The striatum is a brain region that controls movement and some forms of cognition and is most significantly impacted in HD. However, despite well-documented deficits in learning and memory in HD, knowledge of the potential implication of other brain regions such as the hippocampus remains limited. Objective Here, we study the comparative impact of enhanced mHTT aggregation and neuropathology in the striatum and hippocampus of two HD mouse models. Methods We utilized the zQ175 as a control HD mouse model and the Q175DN mice lacking the PGK-Neomycin cassette generated in house. We performed a comparative characterization of the neuropathology between zQ175 and Q175DN mice in the striatum and the hippocampus by assessing HTT aggregation, neuronal and glial pathology, chaperone expression, and synaptic density. Results We showed that Q175DN mice presented enhanced mHTT aggregation in both striatum and hippocampus compared to zQ175. Striatal neurons showed a greater susceptibility to enhanced accumulation of mHTT than hippocampal neurons in Q175DN despite high levels of mHTT in both regions. Contrary to the pathology seen in the striatum, Q175DN hippocampus presented enhanced spare capacity showing increased synaptic density, decreased Iba1+ microglia density and enhanced HSF1 levels in specific subregions of the hippocampus compared to zQ175. Conclusions Q175DN mice are a valuable tool to understand the fundamental susceptibility differences to mHTT toxicity between striatal neurons and other neuronal subtypes. Furthermore, our findings also suggest that cognitive deficits observed in HD animals might arise from either striatum dysfunction or other regions involved in cognitive processes but not from hippocampal degeneration.
Collapse
Affiliation(s)
- Melissa A Solem
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Ross Pelzel
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Nicholas B. Rozema
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Taylor G. Brown
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Emma Reid
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Rachel H. Mansky
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - R Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
2
|
Niso-Santano M, Fuentes JM, Galluzzi L. Immunological aspects of central neurodegeneration. Cell Discov 2024; 10:41. [PMID: 38594240 PMCID: PMC11004155 DOI: 10.1038/s41421-024-00666-z] [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: 12/05/2023] [Accepted: 03/02/2024] [Indexed: 04/11/2024] Open
Abstract
The etiology of various neurodegenerative disorders that mainly affect the central nervous system including (but not limited to) Alzheimer's disease, Parkinson's disease and Huntington's disease has classically been attributed to neuronal defects that culminate with the loss of specific neuronal populations. However, accumulating evidence suggests that numerous immune effector cells and the products thereof (including cytokines and other soluble mediators) have a major impact on the pathogenesis and/or severity of these and other neurodegenerative syndromes. These observations not only add to our understanding of neurodegenerative conditions but also imply that (at least in some cases) therapeutic strategies targeting immune cells or their products may mediate clinically relevant neuroprotective effects. Here, we critically discuss immunological mechanisms of central neurodegeneration and propose potential strategies to correct neurodegeneration-associated immunological dysfunction with therapeutic purposes.
Collapse
Affiliation(s)
- Mireia Niso-Santano
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain.
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain.
| | - José M Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas-Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Steinberg N, Galleguillos D, Zaidi A, Horkey M, Sipione S. Naïve Huntington's disease microglia mount a normal response to inflammatory stimuli but display a partially impaired development of innate immune tolerance that can be counteracted by ganglioside GM1. J Neuroinflammation 2023; 20:276. [PMID: 37996924 PMCID: PMC10668379 DOI: 10.1186/s12974-023-02963-y] [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: 03/07/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023] Open
Abstract
Chronic activation and dysfunction of microglia have been implicated in the pathogenesis and progression of many neurodegenerative disorders, including Huntington's disease (HD). HD is a genetic condition caused by a mutation that affects the folding and function of huntingtin (HTT). Signs of microglia activation have been observed in HD patients even before the onset of symptoms. It is unclear, however, whether pro-inflammatory microglia activation in HD results from cell-autonomous expression of mutant HTT, is the response of microglia to a diseased brain environment, or both. In this study, we used primary microglia isolated from HD knock-in (Q140) and wild-type (Q7) mice to investigate their response to inflammatory conditions in vitro in the absence of confounding effects arising from brain pathology. We show that naïve Q140 microglia do not undergo spontaneous pro-inflammatory activation and respond to inflammatory triggers, including stimulation of TLR4 and TLR2 and exposure to necrotic cells, with similar kinetics of pro-inflammatory gene expression as wild-type microglia. Upon termination of the inflammatory insult, the transcription of pro-inflammatory cytokines is tapered off in Q140 and wild-type microglia with similar kinetics. However, the ability of Q140 microglia to develop tolerance in response to repeated inflammatory stimulations is partially impaired in vitro and in vivo, potentially contributing to the establishment of chronic neuroinflammation in HD. We further show that ganglioside GM1, a glycosphingolipid with anti-inflammatory effects on wild-type microglia, not only decreases the production of pro-inflammatory cytokines and nitric oxide in activated Q140 microglia, but also dramatically dampen microglia response to re-stimulation with LPS in an experimental model of tolerance. These effects are independent from the expression of interleukin 1 receptor associated kinase 3 (Irak-3), a strong modulator of LPS signaling involved in the development of innate immune tolerance and previously shown to be upregulated by immune cell treatment with gangliosides. Altogether, our data suggest that external triggers are required for HD microglia activation, but a cell-autonomous dysfunction that affects the ability of HD microglia to acquire tolerance might contribute to the establishment of neuroinflammation in HD. Administration of GM1 might be beneficial to attenuate chronic microglia activation and neuroinflammation.
Collapse
Affiliation(s)
- Noam Steinberg
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
| | - Danny Galleguillos
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Asifa Zaidi
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada
| | | | - Simonetta Sipione
- Department of Pharmacology, Neuroscience and Mental Health Institute and Glycomics Institute of Alberta, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
5
|
Siew JJ, Chern Y, Khoo KH, Angata T. Roles of Siglecs in neurodegenerative diseases. Mol Aspects Med 2023; 90:101141. [PMID: 36089405 DOI: 10.1016/j.mam.2022.101141] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 02/08/2023]
Abstract
Microglia are resident myeloid cells in the central nervous system (CNS) with a unique developmental origin, playing essential roles in developing and maintaining the CNS environment. Recent studies have revealed the involvement of microglia in neurodegenerative diseases, such as Alzheimer's disease, through the modulation of neuroinflammation. Several members of the Siglec family of sialic acid recognition proteins are expressed on microglia. Since the discovery of the genetic association between a polymorphism in the CD33 gene and late-onset Alzheimer's disease, significant efforts have been made to elucidate the molecular mechanism underlying the association between the polymorphism and Alzheimer's disease. Furthermore, recent studies have revealed additional potential associations between Siglecs and Alzheimer's disease, implying that the reduced signal from inhibitory Siglec may have an overall protective effect in lowering the disease risk. Evidences suggesting the involvement of Siglecs in other neurodegenerative diseases are also emerging. These findings could help us predict the roles of Siglecs in other neurodegenerative diseases. However, little is known about the functionally relevant Siglec ligands in the brain, which represents a new frontier. Understanding how microglial Siglecs and their ligands in CNS contribute to the regulation of CNS homeostasis and pathogenesis of neurodegenerative diseases may provide us with a new avenue for disease prevention and intervention.
Collapse
Affiliation(s)
- Jian Jing Siew
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
6
|
Etxeberria-Rekalde E, Alzola-Aldamizetxebarria S, Flunkert S, Hable I, Daurer M, Neddens J, Hutter-Paier B. Quantification of Huntington's Disease Related Markers in the R6/2 Mouse Model. Front Mol Neurosci 2021; 13:617229. [PMID: 33505246 PMCID: PMC7831778 DOI: 10.3389/fnmol.2020.617229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Huntington’s disease (HD) is caused by an expansion of CAG triplets in the huntingtin gene, leading to severe neuropathological changes that result in a devasting and lethal phenotype. Neurodegeneration in HD begins in the striatum and spreads to other brain regions such as cortex and hippocampus, causing motor and cognitive dysfunctions. To understand the signaling pathways involved in HD, animal models that mimic the human pathology are used. The R6/2 mouse as model of HD was already shown to present major neuropathological changes in the caudate putamen and other brain regions, but recently established biomarkers in HD patients were yet not analyzed in these mice. We therefore performed an in-depth analysis of R6/2 mice to establish new and highly translational readouts focusing on Ctip2 as biological marker for motor system-related neurons and translocator protein (TSPO) as a promising readout for early neuroinflammation. Our results validate already shown pathologies like mutant huntingtin aggregates, ubiquitination, and brain atrophy, but also provide evidence for decreased tyrosine hydroxylase and Ctip2 levels as indicators of a disturbed motor system, while vesicular acetyl choline transporter levels as marker for the cholinergic system barely change. Additionally, increased astrocytosis and activated microglia were observed by GFAP, Iba1 and TSPO labeling, illustrating, that TSPO is a more sensitive marker for early neuroinflammation compared to GFAP and Iba1. Our results thus demonstrate a high sensitivity and translational value of Ctip2 and TSPO as new marker for the preclinical evaluation of new compounds in the R6/2 mouse model of HD.
Collapse
Affiliation(s)
| | | | | | - Isabella Hable
- QPS Austria GmbH, Grambach, Austria.,Department of Health Studies, FH Joanneum University of Applied Sciences, Graz, Austria
| | | | | | | |
Collapse
|
7
|
Savage JC, St-Pierre MK, Carrier M, El Hajj H, Novak SW, Sanchez MG, Cicchetti F, Tremblay MÈ. Microglial physiological properties and interactions with synapses are altered at presymptomatic stages in a mouse model of Huntington's disease pathology. J Neuroinflammation 2020; 17:98. [PMID: 32241286 PMCID: PMC7118932 DOI: 10.1186/s12974-020-01782-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder that affects cognitive and motor abilities by primarily targeting the striatum and cerebral cortex. HD is caused by a mutation elongating the CAG repeats within the Huntingtin gene, resulting in HTT protein misfolding. Although the genetic cause of HD has been established, the specific susceptibility of neurons within various brain structures has remained elusive. Microglia, which are the brain's resident macrophages, have emerged as important players in neurodegeneration. Nevertheless, few studies have examined their implication in HD. METHODS To provide novel insights, we investigated the maturation and dysfunction of striatal microglia using the R6/2 mouse model of HD. This transgenic model, which presents with 120+/-5 CAG repeats, displays progressive motor deficits beginning at 6 weeks of age, with full incapacitation by 13 weeks. We studied microglial morphology, phagocytic capacity, and synaptic contacts in the striatum of R6/2 versus wild-type (WT) littermates at 3, 10, and 13 weeks of age, using a combination of light and transmission electron microscopy. We also reconstructed dendrites and determined synaptic density within the striatum of R6/2 and WT littermates, at nanoscale resolution using focused ion beam scanning electron microscopy. RESULTS At 3 weeks of age, prior to any known motor deficits, microglia in R6/2 animals displayed a more mature morphological phenotype than WT animals. Microglia from R6/2 mice across all ages also demonstrated increased phagocytosis, as revealed by light microscopy and transmission electron microscopy. Furthermore, microglial processes from 10-week-old R6/2 mice made fewer contacts with synaptic structures than microglial processes in 3-week-old R6/2 mice and age-matched WT littermates. Synaptic density was not affected by genotype at 3 weeks of age but increased with maturation in WT mice. The location of synapses was lastly modified in R6/2 mice compared with WT controls, from targeting dendritic spines to dendritic trunks at both 3 and 10 weeks of age. CONCLUSIONS These findings suggest that microglia may play an intimate role in synaptic alteration and loss during HD pathogenesis.
Collapse
Affiliation(s)
- Julie C Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada.
| | - Marie-Kim St-Pierre
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
| | - Micaël Carrier
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
| | - Hassan El Hajj
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
| | - Sammy Weiser Novak
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Maria Gabriela Sanchez
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
| | - Francesca Cicchetti
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada
- Département de psychiatrie et neurosciences, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, Boulevard Laurier, T2-50, Québec, QC, G1V 4G2, Canada.
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
8
|
Yu-Taeger L, Stricker-Shaver J, Arnold K, Bambynek-Dziuk P, Novati A, Singer E, Lourhmati A, Fabian C, Magg J, Riess O, Schwab M, Stolzing A, Danielyan L, Nguyen HHP. Intranasal Administration of Mesenchymal Stem Cells Ameliorates the Abnormal Dopamine Transmission System and Inflammatory Reaction in the R6/2 Mouse Model of Huntington Disease. Cells 2019; 8:E595. [PMID: 31208073 PMCID: PMC6628278 DOI: 10.3390/cells8060595] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022] Open
Abstract
Intrastriatal administration of mesenchymal stem cells (MSCs) has shown beneficial effects in rodent models of Huntington disease (HD). However, the invasive nature of surgical procedure and its potential to trigger the host immune response may limit its clinical use. Hence, we sought to evaluate the non-invasive intranasal administration (INA) of MSC delivery as an effective alternative route in HD. GFP-expressing MSCs derived from bone marrow were intranasally administered to 4-week-old R6/2 HD transgenic mice. MSCs were detected in the olfactory bulb, midbrain and striatum five days post-delivery. Compared to phosphate-buffered saline (PBS)-treated littermates, MSC-treated R6/2 mice showed an increased survival rate and attenuated circadian activity disruption assessed by locomotor activity. MSCs increased the protein expression of DARPP-32 and tyrosine hydroxylase (TH) and downregulated gene expression of inflammatory modulators in the brain 7.5 weeks after INA. While vehicle treated R6/2 mice displayed decreased Iba1 expression and altered microglial morphology in comparison to the wild type littermates, MSCs restored both, Iba1 level and the thickness of microglial processes in the striatum of R6/2 mice. Our results demonstrate significantly ameliorated phenotypes of R6/2 mice after MSCs administration via INA, suggesting this method as an effective delivering route of cells to the brain for HD therapy.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Janice Stricker-Shaver
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Katrin Arnold
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Patrycja Bambynek-Dziuk
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Arianna Novati
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Elisabeth Singer
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Ali Lourhmati
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
| | - Claire Fabian
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Janine Magg
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Matthias Schwab
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, D-70376 Stuttgart, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Alexandra Stolzing
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Hoa Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany.
- Departments of Medical Chemistry and Biochemistry, Yerevan State Medical University, 0025 Yerevan, Armenia.
| |
Collapse
|
9
|
Cohen EM, Mohammed S, Kavurma M, Nedoboy PE, Cartland S, Farnham MM, Pilowsky PM. Microglia in the RVLM of SHR have reduced P2Y12R and CX3CR1 expression, shorter processes, and lower cell density. Auton Neurosci 2019; 216:9-16. [DOI: 10.1016/j.autneu.2018.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023]
|
10
|
Changes in neocortical and hippocampal microglial cells during hibernation. Brain Struct Funct 2017; 223:1881-1895. [PMID: 29260372 DOI: 10.1007/s00429-017-1596-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/15/2017] [Indexed: 12/29/2022]
Abstract
Mammalian hibernation proceeds alongside a wide range of complex brain adaptive changes that appear to protect the brain from extreme hypoxia and hypothermia. Using immunofluorescence, confocal microscopy, quantitative analysis methods and intracellular injections, we have characterized microglia morphological changes that occur in the neocortex and hippocampus of the Syrian hamster during hibernation. In euthermic hamsters, microglial cells showed the typical ramified/resting morphology with multiple long, thin and highly-branched processes homogeneously immunostained for Iba-1. However, during torpor, microglial cell process numbers increase significantly accompanied by a shortening of the Iba-1 immunoreactive processes, which show a fragmented appearance. Adaptative changes of microglial cells during torpor coursed with no expression of microglial cell activation markers. We discuss the possibility that these morphological changes may contribute to neuronal damage prevention during hibernation.
Collapse
|
11
|
Yu-Taeger L, Bonin M, Stricker-Shaver J, Riess O, Nguyen HHP. Dysregulation of gene expression in the striatum of BACHD rats expressing full-length mutant huntingtin and associated abnormalities on molecular and protein levels. Neuropharmacology 2017; 117:260-272. [DOI: 10.1016/j.neuropharm.2017.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/17/2017] [Accepted: 01/27/2017] [Indexed: 11/26/2022]
|
12
|
Coffey SR, Bragg RM, Minnig S, Ament SA, Cantle JP, Glickenhaus A, Shelnut D, Carrillo JM, Shuttleworth DD, Rodier JA, Noguchi K, Bennett CF, Price ND, Kordasiewicz HB, Carroll JB. Peripheral huntingtin silencing does not ameliorate central signs of disease in the B6.HttQ111/+ mouse model of Huntington's disease. PLoS One 2017; 12:e0175968. [PMID: 28453524 PMCID: PMC5409169 DOI: 10.1371/journal.pone.0175968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/03/2017] [Indexed: 01/20/2023] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease whose predominant neuropathological signature is the selective loss of medium spiny neurons in the striatum. Despite this selective neuropathology, the mutant protein (huntingtin) is found in virtually every cell so far studied, and, consequently, phenotypes are observed in a wide range of organ systems both inside and outside the central nervous system. We, and others, have suggested that peripheral dysfunction could contribute to the rate of progression of striatal phenotypes of HD. To test this hypothesis, we lowered levels of huntingtin by treating mice with antisense oligonucleotides (ASOs) targeting the murine Huntingtin gene. To study the relationship between peripheral huntingtin levels and striatal HD phenotypes, we utilized a knock-in model of the human HD mutation (the B6.HttQ111/+ mouse). We treated mice with ASOs from 2-10 months of age, a time period over which significant HD-relevant signs progressively develop in the brains of HttQ111/+ mice. Peripheral treatment with ASOs led to persistent reduction of huntingtin protein in peripheral organs, including liver (64% knockdown), brown adipose (66% knockdown), and white adipose tissues (71% knockdown). This reduction was not associated with alterations in the severity of HD-relevant signs in the striatum of HttQ111/+ mice at the end of the study, including transcriptional dysregulation, the accumulation of neuronal intranuclear inclusions, and behavioral changes such as subtle hypoactivity and reduced exploratory drive. These results suggest that the amount of peripheral reduction achieved in the current study does not significantly impact the progression of HD-relevant signs in the central nervous system.
Collapse
Affiliation(s)
- Sydney R. Coffey
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Robert M. Bragg
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Shawn Minnig
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Seth A. Ament
- Institute for Genome Sciences and Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States of America
- Institute for Systems Biology, Seattle, WA, United States of America
| | - Jeffrey P. Cantle
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Anne Glickenhaus
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Daniel Shelnut
- Department of Mathematics, Western Washington University, Bellingham, WA, United States of America
| | - José M. Carrillo
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Dominic D. Shuttleworth
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| | - Julie-Anne Rodier
- INSERM U1216, Grenoble Institute of Neuroscience, Grenoble, France. Université Grenoble Alpes, Grenoble, France
| | - Kimihiro Noguchi
- Department of Mathematics, Western Washington University, Bellingham, WA, United States of America
| | | | - Nathan D. Price
- Institute for Genome Sciences and Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | | - Jeffrey B. Carroll
- Behavioral Neuroscience Program, Psychology Department, Western Washington University, Bellingham, WA, United States of America
| |
Collapse
|
13
|
Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol Neurobiol 2016; 54:7567-7584. [DOI: 10.1007/s12035-016-0245-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023]
|
14
|
Scheffold A, Holtman IR, Dieni S, Brouwer N, Katz SF, Jebaraj BMC, Kahle PJ, Hengerer B, Lechel A, Stilgenbauer S, Boddeke EWGM, Eggen BJL, Rudolph KL, Biber K. Telomere shortening leads to an acceleration of synucleinopathy and impaired microglia response in a genetic mouse model. Acta Neuropathol Commun 2016; 4:87. [PMID: 27550225 PMCID: PMC4994259 DOI: 10.1186/s40478-016-0364-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease is one of the most common neurodegenerative disorders of the elderly and ageing hence described to be a major risk factor. Telomere shortening as a result of the inability to fully replicate the ends of linear chromosomes is one of the hallmarks of ageing. The role of telomere dysfunction in neurological diseases and the ageing brain is not clarified and there is an ongoing discussion whether telomere shortening is linked to Parkinson’s disease. Here we studied a mouse model of Parkinson’s disease (Thy-1 [A30P] α-synuclein transgenic mouse model) in the background of telomere shortening (Terc knockout mouse model). α-synuclein transgenic mice with short telomeres (αSYNtg/tg G3Terc-/-) developed an accelerated disease with significantly decreased survival. This accelerated phenotype of mice with short telomeres was characterized by a declined motor performance and an increased formation of α-synuclein aggregates. Immunohistochemical analysis and mRNA expression studies revealed that the disease end-stage brain stem microglia showed an impaired response in αSYNtg/tg G3Terc-/- microglia animals. These results provide the first experimental data that telomere shortening accelerates α-synuclein pathology that is linked to limited microglia function in the brainstem.
Collapse
|
15
|
Meunier C, Merienne N, Jollé C, Déglon N, Pellerin L. Astrocytes are key but indirect contributors to the development of the symptomatology and pathophysiology of Huntington's disease. Glia 2016; 64:1841-56. [PMID: 27442486 DOI: 10.1002/glia.23022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/09/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease in which an early and selective vulnerability of striatal Spiny Projection Neurons is observed. However, several studies have highlighted the implication of glial cells, and in particular astrocytes, in the pathophysiological mechanisms of this disease. A better understanding of the respective contributions of neurons and astrocytes in HD is needed and would be important for the development of new therapeutic approaches. Today, no comparable in vivo models expressing the mutant HTT selectively in astrocytes or in neurons are available. In this study, we developed comparable cell-type specific mouse models expressing a fragment of Huntingtin specifically in neurons, astrocytes, or in both cell populations of the adult mouse basal ganglia circuit. This approach allowed us to characterize behavioral alterations occurring as soon as 4 weeks postinjection. Interestingly, less severe but significant behavioral alterations were also observed in the two cell-type specific models. We further showed that astrocytes are less affected by mHTT compared to neurons, in particular concerning mHTT aggregation. Additionally, a more indirect contribution of astrocytes compared to neurons was observed in several pathophysiological mechanisms such as astrogliosis and neuronal dysfunction. Finally, we showed that direct and indirect transcriptional alterations within the glial glutamatergic clearing system are caused by astrocytic and neuronal expression of mHTT, respectively. We anticipate that our study will help to better understand the contributions of astrocytes to HD and guide future therapeutic efforts. GLIA 2016;64:1841-1856.
Collapse
Affiliation(s)
- Cécile Meunier
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Merienne
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies (LCMN), Lausanne University Hospital, Lausanne, Switzerland.,Neuroscience Research Center (CRN), LCMN, Lausanne University Hospital, Lausanne, Switzerland
| | - Charlotte Jollé
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Nicole Déglon
- Department of Clinical Neurosciences, Laboratory of Cellular and Molecular Neurotherapies (LCMN), Lausanne University Hospital, Lausanne, Switzerland.,Neuroscience Research Center (CRN), LCMN, Lausanne University Hospital, Lausanne, Switzerland
| | - Luc Pellerin
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
16
|
Tischer J, Krueger M, Mueller W, Staszewski O, Prinz M, Streit WJ, Bechmann I. Inhomogeneous distribution of Iba-1 characterizes microglial pathology in Alzheimer's disease. Glia 2016; 64:1562-72. [PMID: 27404378 DOI: 10.1002/glia.23024] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/12/2016] [Accepted: 06/20/2016] [Indexed: 11/06/2022]
Abstract
Microglial dystrophy has recently been described as a morphological phenotype of microglia that differs from resting and activated states by spheroid formation and cytorrhexis. In thick sections immunolabeled for HLA-DR or Iba-1 dystrophic microglial processes lose their typical, homogeneous staining pattern and appear to be fragmented or clustered. In this study, we performed double immunofluorescence and electron microscopy to determine if this labeling pattern indeed reflects complete separation of microglial processes from the soma. Using Iba-1/CD68 and Iba-1/MHC class II, as microglial markers, we observed that isolated Iba-1 fragments were still connected to each other by segments of the microglial process immune positive for CD68 or MHC class II. Ultrathin serial sections of two Iba-1 fragments which appeared to be disconnected from each other at the light microscopical level revealed a still existing "bridge" with a diameter of around 0.182 µm. Therefore, microglial dystrophy may reflect alterations of the cytoskeleton ultimately leading to slow cytorrhexis. GLIA 2016;64:1562-1572.
Collapse
Affiliation(s)
- Jasmin Tischer
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Wolf Mueller
- Department of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Ori Staszewski
- Neurozentrum, Institute of Neuropathology, Freiburg, Germany
| | - Marco Prinz
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Wolfgang J Streit
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
17
|
Lemon JA, Aksenov V, Samigullina R, Aksenov S, Rodgers WH, Rollo CD, Boreham DR. A multi-ingredient dietary supplement abolishes large-scale brain cell loss, improves sensory function, and prevents neuronal atrophy in aging mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:382-404. [PMID: 27199101 DOI: 10.1002/em.22019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/14/2016] [Indexed: 06/05/2023]
Abstract
Transgenic growth hormone mice (TGM) are a recognized model of accelerated aging with characteristics including chronic oxidative stress, reduced longevity, mitochondrial dysfunction, insulin resistance, muscle wasting, and elevated inflammatory processes. Growth hormone/IGF-1 activate the Target of Rapamycin known to promote aging. TGM particularly express severe cognitive decline. We previously reported that a multi-ingredient dietary supplement (MDS) designed to offset five mechanisms associated with aging extended longevity, ameliorated cognitive deterioration and significantly reduced age-related physical deterioration in both normal mice and TGM. Here we report that TGM lose more than 50% of cells in midbrain regions, including the cerebellum and olfactory bulb. This is comparable to severe Alzheimer's disease and likely explains their striking age-related cognitive impairment. We also demonstrate that the MDS completely abrogates this severe brain cell loss, reverses cognitive decline and augments sensory and motor function in aged mice. Additionally, histological examination of retinal structure revealed markers consistent with higher numbers of photoreceptor cells in aging and supplemented mice. We know of no other treatment with such efficacy, highlighting the potential for prevention or amelioration of human neuropathologies that are similarly associated with oxidative stress, inflammation and cellular dysfunction. Environ. Mol. Mutagen. 57:382-404, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- J A Lemon
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street West, Hamilton ON, Canada, L8S 4K1
| | - V Aksenov
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton ON, Canada, L8S 4K1
| | - R Samigullina
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton ON, Canada, L8S 4K1
| | - S Aksenov
- Department of Pathology, New York-Presbyterian/Queens Hospital, 56-45 Main Street, Flushing, New York, 11355
| | - W H Rodgers
- Department of Pathology, New York-Presbyterian/Queens Hospital, 56-45 Main Street, Flushing, New York, 11355
| | - C D Rollo
- Department of Biology, McMaster University, 1280 Main Street West, Hamilton ON, Canada, L8S 4K1
| | - D R Boreham
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street West, Hamilton ON, Canada, L8S 4K1
- Medical Sciences Division, Northern Ontario School of Medicine, 935 Ramsey Lake Road, Sudbury ON, Canada, P3E 2C6
| |
Collapse
|
18
|
Alto LT, Chen X, Ruhn KA, Treviño I, Tansey MG. AAV-dominant negative tumor necrosis factor (DN-TNF) gene transfer to the striatum does not rescue medium spiny neurons in the YAC128 mouse model of Huntington's disease. PLoS One 2014; 9:e96544. [PMID: 24824433 PMCID: PMC4019512 DOI: 10.1371/journal.pone.0096544] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/08/2014] [Indexed: 11/19/2022] Open
Abstract
CNS inflammation is a hallmark of neurodegenerative disease, and recent studies suggest that the inflammatory response may contribute to neuronal demise. In particular, increased tumor necrosis factor (TNF) signaling is implicated in the pathology of both Parkinson's disease (PD) and Alzheimer's disease (AD). We have previously shown that localized gene delivery of dominant negative TNF to the degenerating brain region can limit pathology in animal models of PD and AD. TNF is upregulated in Huntington's disease (HD), like in PD and AD, but it is unknown whether TNF signaling contributes to neuronal degeneration in HD. We used in vivo gene delivery to test whether selective reduction of soluble TNF signaling could attenuate medium spiny neuron (MSN) degeneration in the YAC128 transgenic (TG) mouse model of Huntington's disease (HD). AAV vectors encoding cDNA for dominant-negative tumor necrosis factor (DN-TNF) or GFP (control) were injected into the striatum of young adult wild type WT and YAC128 TG mice and achieved 30-50% target coverage. Expression of dominant negative TNF protein was confirmed immunohistologically and biochemically and was maintained as mice aged to one year, but declined significantly over time. However, the extent of striatal DN-TNF gene transfer achieved in our studies was not sufficient to achieve robust effects on neuroinflammation, rescue degenerating MSNs or improve motor function in treated mice. Our findings suggest that alternative drug delivery strategies should be explored to determine whether greater target coverage by DN-TNF protein might afford some level of neuroprotection against HD-like pathology and/or that soluble TNF signaling may not be the primary driver of striatal neuroinflammation and MSN loss in YAC128 TG mice.
Collapse
Affiliation(s)
- Laura Taylor Alto
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xi Chen
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly A. Ruhn
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Isaac Treviño
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Malú G. Tansey
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Biber K, Owens T, Boddeke E. What is microglia neurotoxicity (Not)? Glia 2014; 62:841-54. [PMID: 24590682 DOI: 10.1002/glia.22654] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/10/2014] [Accepted: 02/14/2014] [Indexed: 01/19/2023]
Abstract
Microglia most likely appeared early in evolution as they are not only present in vertebrates, but are also found in nervous systems of various nonvertebrate organisms. Mammalian microglia are derived from a specific embryonic, self-renewable myeloid cell population that is throughout lifetime not replaced by peripheral myeloid cells. These phylogenic and ontogenic features suggest that microglia serve vital functions. Yet, microglia often are described as neurotoxic cells, that actively kill (healthy) neurons. Since it is from an evolutionary point of view difficult to understand why an important and vulnerable organ like the brain should host numerous potential killers, we here review the concept of microglia neurotoxicity. On one hand it is discussed that most of our understanding about how microglia kill neurons is based on in vitro experiments or correlative staining studies that suffer from the difficulty to discriminate microglia and peripheral myeloid cells in the diseased brain. On the other hand it is described that a more functional approach by mutating, inactivating or deleting microglia is seldom associated with a beneficial outcome in an acute injury situation, suggesting that microglia are normally important protective elements in the brain. This might change in chronic disease or the aged brain, where; however, it remains to be established whether microglia simply lose their protective capacities or whether microglia become truly neurotoxic cells.
Collapse
Affiliation(s)
- Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Hauptstrasse 5, 79104, Freiburg, Germany; Department of Neuroscience, University Medical Center Groningen, Ant. Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | | | | |
Collapse
|
20
|
Maheshwari M, Bhutani S, Das A, Mukherjee R, Sharma A, Kino Y, Nukina N, Jana NR. Dexamethasone induces heat shock response and slows down disease progression in mouse and fly models of Huntington's disease. Hum Mol Genet 2013; 23:2737-51. [PMID: 24381308 DOI: 10.1093/hmg/ddt667] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by abnormal expansion of glutamine repeats in the protein huntingtin. In HD brain, mutant huntingtin undergoes proteolytic processing, and its N-terminal fragment containing poly-glutamine repeats accumulate as insoluble aggregates leading to the defect in cellular protein quality control system and heat shock response (HSR). Here we demonstrate that the defective HSR in the brain is due to the down-regulation of heat shock factor 1 (HSF1) in both mice and fly models of HD. Interestingly, treatment of dexamethasone (a synthetic glucocorticoid) to HD mice or flies significantly increased the expression and transactivation of HSF1 and induction of HSR and these effects are mediated through the down-regulation of HSP90. Dexamethasone treatment also significantly decreased the aggregate load and transient recovery of HD-related behavioural phenotypes in both disease models. These results suggest that dexamethasone could be a potential therapeutic molecule for the treatment of HD and related poly-glutamine disorders.
Collapse
Affiliation(s)
- Megha Maheshwari
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122 050, India
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Rattray I, Smith EJ, Crum WR, Walker TA, Gale R, Bates GP, Modo M. Correlations of behavioral deficits with brain pathology assessed through longitudinal MRI and histopathology in the R6/1 mouse model of Huntington's disease. PLoS One 2013; 8:e84726. [PMID: 24367693 PMCID: PMC3868608 DOI: 10.1371/journal.pone.0084726] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 11/18/2013] [Indexed: 02/04/2023] Open
Abstract
Huntington's disease (HD) is caused by the expansion of a CAG repeat in the huntingtin (HTT) gene. The R6 mouse models of HD express a mutant version of exon 1 HTT and typically develop motor and cognitive impairments, a widespread huntingtin (HTT) aggregate pathology and brain atrophy. Unlike the more commonly used R6/2 mouse line, R6/1 mice have fewer CAG repeats and, subsequently, a less rapid pathological decline. Compared to the R6/2 line, fewer descriptions of the progressive pathologies exhibited by R6/1 mice exist. The association between the molecular and cellular neuropathology with brain atrophy, and with the development of behavioral phenotypes remains poorly understood in many models of HD. In attempt to link these factors in the R6/1 mouse line, we have performed detailed assessments of behavior and of regional brain abnormalities determined through longitudinal, in vivo magnetic resonance imaging (MRI), as well as an end-stage, ex vivo MRI study and histological assessment. We found progressive decline in both motor and non-motor related behavioral tasks in R6/1 mice, first evident at 11 weeks of age. Regional brain volumes were generally unaffected at 9 weeks, but by 17 weeks there was significant grey matter atrophy. This age-related brain volume loss was validated using a more precise, semi-automated Tensor Based morphometry assessment. As well as these clear progressive phenotypes, mutant HTT (mHTT) protein, the hallmark of HD molecular pathology, was widely distributed throughout the R6/1 brain and was accompanied by neuronal loss. Despite these seemingly concomitant, robust pathological phenotypes, there appeared to be little correlation between the three main outcome measures: behavioral performance, MRI-detected brain atrophy and histopathology. In conclusion, R6/1 mice exhibit many features of HD, but the underlying mechanisms driving these clear behavioral disturbances and the brain volume loss, still remain unclear.
Collapse
Affiliation(s)
- Ivan Rattray
- King’s College London, Institute of Psychiatry, Department of Neuroscience, London, United Kingdom
- King’s College London, Department of Medical and Molecular Genetics, London, United Kingdom
| | - Edward J. Smith
- King’s College London, Institute of Psychiatry, Department of Neuroscience, London, United Kingdom
- King’s College London, Department of Medical and Molecular Genetics, London, United Kingdom
| | - William R. Crum
- King’s College London, Department of Neuroimaging, Institute of Psychiatry, London, United Kingdom
| | - Thomas A. Walker
- King’s College London, Department of Medical and Molecular Genetics, London, United Kingdom
| | - Richard Gale
- King’s College London, Department of Medical and Molecular Genetics, London, United Kingdom
| | - Gillian P. Bates
- King’s College London, Department of Medical and Molecular Genetics, London, United Kingdom
| | - Michel Modo
- King’s College London, Institute of Psychiatry, Department of Neuroscience, London, United Kingdom
- University of Pittsburgh, Department of Radiology, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
22
|
Correlations of behavioral deficits with brain pathology assessed through longitudinal MRI and histopathology in the R6/2 mouse model of HD. PLoS One 2013; 8:e60012. [PMID: 23593159 PMCID: PMC3617160 DOI: 10.1371/journal.pone.0060012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 02/20/2013] [Indexed: 11/30/2022] Open
Abstract
Huntington’s disease (HD) is caused by the expansion of a CAG repeat in the huntingtin (HTT) gene. The R6/2 mouse model of HD expresses a mutant version of exon 1 HTT and develops motor and cognitive impairments, a widespread huntingtin (HTT) aggregate pathology and brain atrophy. Despite the vast number of studies that have been performed on this model, the association between the molecular and cellular neuropathology with brain atrophy, and with the development of behavioral phenotypes remains poorly understood. In an attempt to link these factors, we have performed longitudinal assessments of behavior (rotarod, open field, passive avoidance) and of regional brain abnormalities determined through magnetic resonance imaging (MRI) (whole brain, striatum, cortex, hippocampus, corpus callosum), as well as an end-stage histological assessment. Detailed correlative analyses of these three measures were then performed. We found a gender-dependent emergence of motor impairments that was associated with an age-related loss of regional brain volumes. MRI measurements further indicated that there was no striatal atrophy, but rather a lack of striatal growth beyond 8 weeks of age. T2 relaxivity further indicated tissue-level changes within brain regions. Despite these dramatic motor and neuroanatomical abnormalities, R6/2 mice did not exhibit neuronal loss in the striatum or motor cortex, although there was a significant increase in neuronal density due to tissue atrophy. The deposition of the mutant HTT (mHTT) protein, the hallmark of HD molecular pathology, was widely distributed throughout the brain. End-stage histopathological assessments were not found to be as robustly correlated with the longitudinal measures of brain atrophy or motor impairments. In conclusion, modeling pre-manifest and early progression of the disease in more slowly progressing animal models will be key to establishing which changes are causally related.
Collapse
|
23
|
Kwan W, Träger U, Davalos D, Chou A, Bouchard J, Andre R, Miller A, Weiss A, Giorgini F, Cheah C, Möller T, Stella N, Akassoglou K, Tabrizi SJ, Muchowski PJ. Mutant huntingtin impairs immune cell migration in Huntington disease. J Clin Invest 2012; 122:4737-47. [PMID: 23160193 DOI: 10.1172/jci64484] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/27/2012] [Indexed: 11/17/2022] Open
Abstract
In Huntington disease (HD), immune cells are activated before symptoms arise; however, it is unclear how the expression of mutant huntingtin (htt) compromises the normal functions of immune cells. Here we report that primary microglia from early postnatal HD mice were profoundly impaired in their migration to chemotactic stimuli, and expression of a mutant htt fragment in microglial cell lines was sufficient to reproduce these deficits. Microglia expressing mutant htt had a retarded response to a laser-induced brain injury in vivo. Leukocyte recruitment was defective upon induction of peritonitis in HD mice at early disease stages and was normalized upon genetic deletion of mutant htt in immune cells. Migration was also strongly impaired in peripheral immune cells from pre-manifest human HD patients. Defective actin remodeling in immune cells expressing mutant htt likely contributed to their migration deficit. Our results suggest that these functional changes may contribute to immune dysfunction and neurodegeneration in HD, and may have implications for other polyglutamine expansion diseases in which mutant proteins are ubiquitously expressed.
Collapse
Affiliation(s)
- Wanda Kwan
- Biomedical Sciences Program, UCSF, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Figiel M, Szlachcic WJ, Switonski PM, Gabka A, Krzyzosiak WJ. Mouse models of polyglutamine diseases: review and data table. Part I. Mol Neurobiol 2012; 46:393-429. [PMID: 22956270 PMCID: PMC3461215 DOI: 10.1007/s12035-012-8315-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/29/2012] [Indexed: 12/23/2022]
Abstract
Polyglutamine (polyQ) disorders share many similarities, such as a common mutation type in unrelated human causative genes, neurological character, and certain aspects of pathogenesis, including morphological and physiological neuronal alterations. The similarities in pathogenesis have been confirmed by findings that some experimental in vivo therapy approaches are effective in multiple models of polyQ disorders. Additionally, mouse models of polyQ diseases are often highly similar between diseases with respect to behavior and the features of the disease. The common features shared by polyQ mouse models may facilitate the investigation of polyQ disorders and may help researchers explore the mechanisms of these diseases in a broader context. To provide this context and to promote the understanding of polyQ disorders, we have collected and analyzed research data about the characterization and treatment of mouse models of polyQ diseases and organized them into two complementary Excel data tables. The data table that is presented in this review (Part I) covers the behavioral, molecular, cellular, and anatomic characteristics of polyQ mice and contains the most current knowledge about polyQ mouse models. The structure of this data table is designed in such a way that it can be filtered to allow for the immediate retrieval of the data corresponding to a single mouse model or to compare the shared and unique aspects of many polyQ models. The second data table, which is presented in another publication (Part II), covers therapeutic research in mouse models by summarizing all of the therapeutic strategies employed in the treatment of polyQ disorders, phenotypes that are used to examine the effects of the therapy, and therapeutic outcomes.
Collapse
Affiliation(s)
- Maciej Figiel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland.
| | | | | | | | | |
Collapse
|
25
|
An in vitro perspective on the molecular mechanisms underlying mutant huntingtin protein toxicity. Cell Death Dis 2012; 3:e382. [PMID: 22932724 PMCID: PMC3434668 DOI: 10.1038/cddis.2012.121] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder whose main hallmark is brain atrophy. However, several peripheral organs are considerably affected and their symptoms may, in fact, manifest before those resulting from brain pathology. HD is of genetic origin and caused by a mutation in the huntingtin gene. The mutated protein has detrimental effects on cell survival, but whether the mutation leads to a gain of toxic function or a loss of function of the altered protein is still highly controversial. Most currently used in vitro models have been designed, to a large extent, to investigate the effects of the aggregation process in neuronal-like cells. However, as the pathology involves several other organs, new in vitro models are critically needed to take into account the deleterious effects of mutant huntingtin in peripheral tissues, and thus to identify new targets that could lead to more effective clinical interventions in the early course of the disease. This review aims to present current in vitro models of HD pathology and to discuss the knowledge that has been gained from these studies as well as the new in vitro tools that have been developed, which should reflect the more global view that we now have of the disease.
Collapse
|
26
|
El-Akabawy G, Rattray I, Johansson SM, Gale R, Bates G, Modo M. Implantation of undifferentiated and pre-differentiated human neural stem cells in the R6/2 transgenic mouse model of Huntington's disease. BMC Neurosci 2012; 13:97. [PMID: 22876937 PMCID: PMC3502570 DOI: 10.1186/1471-2202-13-97] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 07/24/2012] [Indexed: 01/15/2023] Open
Abstract
Background Cell therapy is a potential therapeutic approach for several neurodegenetative disease, including Huntington Disease (HD). To evaluate the putative efficacy of cell therapy in HD, most studies have used excitotoxic animal models with only a few studies having been conducted in genetic animal models. Genetically modified animals should provide a more accurate representation of human HD, as they emulate the genetic basis of its etiology. Results In this study, we aimed to assess the therapeutic potential of a human striatal neural stem cell line (STROC05) implanted in the R6/2 transgenic mouse model of HD. As DARPP-32 GABAergic output neurons are predominately lost in HD, STROC05 cells were also pre-differentiated using purmorphamine, a hedgehog agonist, to yield a greater number of DARPP-32 cells. A bilateral injection of 4.5x105 cells of either undifferentiated or pre-differentiated DARPP-32 cells, however, did not affect outcome compared to a vehicle control injection. Both survival and neuronal differentiation remained poor with a mean of only 161 and 81 cells surviving in the undifferentiated and differentiated conditions respectively. Only a few cells expressed the neuronal marker Fox3. Conclusions Although the rapid brain atrophy and short life-span of the R6/2 model constitute adverse conditions to detect potentially delayed treatment effects, significant technical hurdles, such as poor cell survival and differentiation, were also sub-optimal. Further consideration of these aspects is therefore needed in more enduring transgenic HD models to provide a definite assessment of this cell line’s therapeutic relevance. However, a combination of treatments is likely needed to affect outcome in transgenic models of HD.
Collapse
Affiliation(s)
- Gehan El-Akabawy
- Department of Neuroscience, King's College London, Institute of Psychiatry, London, SE5 9NU, United Kingdom
| | | | | | | | | | | |
Collapse
|
27
|
Lima IVDA, Bastos LFS, Limborço-Filho M, Fiebich BL, de Oliveira ACP. Role of prostaglandins in neuroinflammatory and neurodegenerative diseases. Mediators Inflamm 2012; 2012:946813. [PMID: 22778499 PMCID: PMC3385693 DOI: 10.1155/2012/946813] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/05/2012] [Indexed: 11/17/2022] Open
Abstract
Increasing data demonstrates that inflammation participates in the pathophysiology of neurodegenerative diseases. Among the different inflammatory mediators involved, prostaglandins play an important role. The effects induced by prostaglandins might be mediated by activation of their known receptors or by nonclassical mechanisms. In the present paper, we discuss the evidences that link prostaglandins, as well as the enzymes that produce them, to some neurological diseases.
Collapse
Affiliation(s)
- Isabel Vieira de Assis Lima
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Leandro Francisco Silva Bastos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
- Department of Psychology and Neuroscience, Muenzinger Building, Colorado University of Colorado Boulder, Avenida, Boulder, CO 80309-0354, USA
| | - Marcelo Limborço-Filho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
| | - Bernd L. Fiebich
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
- VivaCell Biotechnology GmbH, Ferdinand-Porsche-Straße 5, 79211 Denzlingen, Germany
| | - Antonio Carlos Pinheiro de Oliveira
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
| |
Collapse
|
28
|
Phillips RJ, Powley TL. Macrophages associated with the intrinsic and extrinsic autonomic innervation of the rat gastrointestinal tract. Auton Neurosci 2012; 169:12-27. [PMID: 22436622 DOI: 10.1016/j.autneu.2012.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 12/28/2022]
Abstract
Interactions between macrophages and the autonomic innervation of gastrointestinal (GI) tract smooth muscle have received little experimental attention. To better understand this relationship, immunohistochemistry was performed on GI whole mounts from rats at three ages. The phenotypes, morphologies, and distributions of gut macrophages are consistent with the cells performing extensive housekeeping functions in the smooth muscle layers. Specifically, a dense population of macrophages was located throughout the muscle wall where they were distributed among the muscle fibers and along the vasculature. Macrophages were also associated with ganglia and connectives of the myenteric plexus and with the sympathetic innervation. Additionally, these cells were in tight registration with the dendrites and axons of the myenteric neurons as well as the varicosities along the length of the sympathetic axons, suggestive of a contribution by the macrophages to the homeostasis of both synapses and contacts between the various elements of the enteric circuitry. Similarly, macrophages were involved in the presumed elimination of neuropathies as indicated by their association with dystrophic neurons and neurites which are located throughout the myenteric plexus and smooth muscle wall of aged rats. Importantly, the patterns of macrophage-neuron interactions in the gut paralleled the much more extensively characterized interactions of macrophages (i.e., microglia) and neurons in the CNS. The present observations in the PNS as well as extrapolations from homologous microglia in the CNS suggest that GI macrophages play significant roles in maintaining the nervous system of the gut in the face of wear and tear, disease, and aging.
Collapse
Affiliation(s)
- Robert J Phillips
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana 47907-2081, USA
| | | |
Collapse
|
29
|
Kraft AD, Kaltenbach LS, Lo DC, Harry GJ. Activated microglia proliferate at neurites of mutant huntingtin-expressing neurons. Neurobiol Aging 2011; 33:621.e17-33. [PMID: 21482444 DOI: 10.1016/j.neurobiolaging.2011.02.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 02/01/2011] [Accepted: 02/16/2011] [Indexed: 01/13/2023]
Abstract
In Huntington's disease (HD), mutated huntingtin (mhtt) causes striatal neurodegeneration which is paralleled by elevated microglia cell numbers. In vitro corticostriatal slice and primary neuronal culture models, in which neuronal expression of mhtt fragments drives HD-like neurotoxicity, were employed to examine wild type microglia during both the initiation and progression of neuronal pathology. As neuronal pathology progressed, microglia initially localized in the vicinity of neurons expressing mhtt fragments increased in number, demonstrated morphological evidence of activation, and expressed the proliferation marker, Ki67. These microglia were positioned along irregular neurites, but did not localize with mhtt inclusions nor exacerbate mhtt fragment-induced neurotoxicity. Prior to neuronal pathology, microglia upregulated ionized calcium binding adaptor molecule 1 (Iba1), signaling a functional shift. With neurodegeneration, interleukin-6 and complement component 1q were increased. The results suggest a stimulatory, proliferative signal for microglia present at the onset of mhtt fragment-induced neurodegeneration. Thus, microglia effect a localized inflammatory response to neuronal mhtt expression that may serve to direct microglial removal of dysfunctional neurites or aberrant synapses, as is required for reparative actions in vivo.
Collapse
Affiliation(s)
- Andrew D Kraft
- Neurotoxicology Group, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | | | |
Collapse
|
30
|
Damani MR, Zhao L, Fontainhas AM, Amaral J, Fariss RN, Wong WT. Age-related alterations in the dynamic behavior of microglia. Aging Cell 2011; 10:263-76. [PMID: 21108733 DOI: 10.1111/j.1474-9726.2010.00660.x] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Microglia, the primary resident immune cells of the central nervous system (CNS), exhibit dynamic behavior involving rapid process motility and cellular migration that is thought to underlie key functions of immune surveillance and tissue repair. Although age-related changes in microglial activation have been implicated in the pathogenesis of neurodegenerative diseases of aging, how dynamic behavior in microglia is influenced by aging is not fully understood. In this study, we employed live imaging of retinal microglia in situ to compare microglial morphology and behavioral dynamics in young and aged animals. We found that aged microglia in the resting state have significantly smaller and less branched dendritic arbors, and also slower process motilities, which probably compromise their ability to survey and interact with their environment continuously. We also found that dynamic microglial responses to injury were age-dependent. While young microglia responded to extracellular ATP, an injury-associated signal, by increasing their motility and becoming more ramified, aged microglia exhibited a contrary response, becoming less dynamic and ramified. In response to laser-induced focal tissue injury, aged microglia demonstrated slower acute responses with lower rates of process motility and cellular migration compared with young microglia. Interestingly, the longer term response of disaggregation from the injury site was retarded in aged microglia, indicating that senescent microglial responses, while slower to initiate, are more sustained. Together, these altered features of microglial behavior at rest and following injury reveal an age-dependent dysregulation of immune response in the CNS that may illuminate microglial contributions to age-related neuroinflammatory degeneration.
Collapse
Affiliation(s)
- Mausam R Damani
- Unit on Neuron-Glia Interactions in Retinal Disease Mechanisms of Disease Section Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
31
|
Sathyasaikumar KV, Stachowski EK, Amori L, Guidetti P, Muchowski PJ, Schwarcz R. Dysfunctional kynurenine pathway metabolism in the R6/2 mouse model of Huntington's disease. J Neurochem 2010; 113:1416-25. [PMID: 20236387 DOI: 10.1111/j.1471-4159.2010.06675.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Elevated concentrations of neurotoxic metabolites of the kynurenine pathway (KP) of tryptophan degradation may play a causative role in Huntington's disease (HD). The brain levels of one of these compounds, 3-hydroxykynurenine (3-HK), are increased in both HD and several mouse models of the disease. In the present study, we examined this impairment in greater detail using the R6/2 mouse, a well-established animal model of HD. Initially, mutant and age-matched wild-type mice received an intrastriatal injection of (3)H-tryptophan to assess the acute, local de novo production of kynurenine, the immediate bioprecursor of 3-HK, in vivo. No effect of genotype was observed between 4 and 12 weeks of age. In contrast, intrastriatally applied (3)H-kynurenine resulted in significantly increased neosynthesis of (3)H-3-HK, but not other tritiated KP metabolites, in the R6/2 striatum. Subsequent ex vivo studies in striatal, cortical and cerebellar tissue revealed substantial increases in the activity of the biosynthetic enzyme of 3-HK, kynurenine 3-monooxygenase and significant reductions in the activity of its degradative enzyme, kynureninase, in HD mice starting at 4 weeks of age. Decreased kynureninase activity was most evident in the cortex and preceded the increase in kynurenine 3-monooxygenase activity. The activity of other KP enzymes showed no consistent brain abnormalities in the mutant mice. These findings suggest that impairments in its immediate metabolic enzymes jointly account for the abnormally high brain levels of 3-HK in the R6/2 model of HD.
Collapse
Affiliation(s)
- Korrapati V Sathyasaikumar
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | | | | | | | | | | |
Collapse
|
32
|
Schwarcz R, Guidetti P, Sathyasaikumar KV, Muchowski PJ. Of mice, rats and men: Revisiting the quinolinic acid hypothesis of Huntington's disease. Prog Neurobiol 2010; 90:230-45. [PMID: 19394403 PMCID: PMC2829333 DOI: 10.1016/j.pneurobio.2009.04.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/17/2009] [Indexed: 12/31/2022]
Abstract
The neurodegenerative disease Huntington's disease (HD) is caused by an expanded polyglutamine (polyQ) tract in the protein huntingtin (htt). Although the gene encoding htt was identified and cloned more than 15 years ago, and in spite of impressive efforts to unravel the mechanism(s) by which mutant htt induces nerve cell death, these studies have so far not led to a good understanding of pathophysiology or an effective therapy. Set against a historical background, we review data supporting the idea that metabolites of the kynurenine pathway (KP) of tryptophan degradation provide a critical link between mutant htt and the pathophysiology of HD. New studies in HD brain and genetic model organisms suggest that the disease may in fact be causally related to early abnormalities in KP metabolism, favoring the formation of two neurotoxic metabolites, 3-hydroxykynurenine and quinolinic acid, over the related neuroprotective agent kynurenic acid. These findings not only link the excitotoxic hypothesis of HD pathology to an impairment of the KP but also define new drug targets and therefore have direct therapeutic implications. Thus, pharmacological normalization of the imbalance in brain KP metabolism may provide clinical benefits, which could be especially effective in early stages of the disease.
Collapse
Affiliation(s)
- Robert Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
33
|
Schwab C, Klegeris A, McGeer PL. Inflammation in transgenic mouse models of neurodegenerative disorders. Biochim Biophys Acta Mol Basis Dis 2009; 1802:889-902. [PMID: 19883753 DOI: 10.1016/j.bbadis.2009.10.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 10/09/2009] [Accepted: 10/23/2009] [Indexed: 12/31/2022]
Abstract
Much evidence is available that inflammation contributes to the development of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Our review investigates how well current mouse models reflect this aspect of the pathogenesis. Transgenic models of AD have been available for several years and are the most extensively studied. Modulation of cytokine levels, activation of microglia and, to a lesser extent, activation of the complement system have been reported. Mouse models of PD and HD so far show less evidence for the involvement of inflammation. An increasing number of transgenic mouse strains is being created to model human neurodegenerative diseases. A perfect model should reflect all aspects of a disease. It is important to evaluate continuously the models for their match with the human disease and reevaluate them in light of new findings in human patients. Although none of the transgenic mouse models recapitulates all aspects of the human disorder they represent, all models have provided valuable information on basic molecular pathways. In particular, the mouse models of Alzheimer disease have also led to the development of new therapeutic strategies such as vaccination and modulation of microglial activity.
Collapse
Affiliation(s)
- Claudia Schwab
- Department of Psychiatry, Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T1Z3, Canada.
| | | | | |
Collapse
|
34
|
Godavarthi SK, Narender D, Mishra A, Goswami A, Rao SNR, Nukina N, Jana NR. Induction of chemokines, MCP-1, and KC in the mutant huntingtin expressing neuronal cells because of proteasomal dysfunction. J Neurochem 2009; 108:787-95. [PMID: 19187096 DOI: 10.1111/j.1471-4159.2008.05823.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Huntington's disease is a hereditary neurodegenerative disorder caused by an aberrant polyglutamine expansion in the amino terminus of the huntingtin protein. The resultant mutant huntingtin form aggregates in neurons and causes neuronal dysfunction and degeneration in many ways including transcriptional dysregulation. Here, we report that the expression of mutant huntingtin in the mouse neuroblastoma cell results in massive transcriptional induction of several chemokines including monocyte chemoattractant protein-1 (MCP-1) and murine chemokine (KC). The mutant huntingtin expressing cells also exhibit proteasomal dysfunction and down-regulation of NF-kappaB activity in a time-dependent manner and both these phenomena regulate the expression of MCP-1 and KC. The expression of MCP-1 and KC are increased in the mutant huntingtin expressing cells in response to mild proteasome inhibition. However, the expression of MCP-1 and KC and proteasome activity are not altered and inflammation is rarely observed in the brain of 12-week-old Huntington's disease transgenic mice in comparison with their age-matched controls. Our result suggests that the mutant huntingtin-induced proteasomal dysfunction can up-regulate the expression of MCP-1 and KC in the neuronal cells and therefore might trigger the inflammation process.
Collapse
Affiliation(s)
- Swetha K Godavarthi
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon, India
| | | | | | | | | | | | | |
Collapse
|
35
|
Lopes KO, Sparks DL, Streit WJ. Microglial dystrophy in the aged and Alzheimer's disease brain is associated with ferritin immunoreactivity. Glia 2008; 56:1048-60. [PMID: 18442088 DOI: 10.1002/glia.20678] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Degeneration of microglial cells may be important for understanding the pathogenesis of aging-related neurodegeneration and neurodegenerative diseases. In this study, we analyzed the morphological characteristics of microglial cells in the nondemented and Alzheimer's disease (AD) human brain using ferritin immunohistochemistry. The central hypothesis was that expression of the iron storage protein ferritin increases the susceptibility of microglia to degeneration, particularly in the aged brain since senescent microglia might become less efficient in maintaining iron homeostasis and free iron can promote oxidative damage. In a primary set of 24 subjects (age range 34-97 years) examined, microglial cells immunoreactive for ferritin were found to constitute a subpopulation of the larger microglial pool labeled with an antibody for HLA-DR antigens. The majority of these ferritin-positive microglia exhibited aberrant morphological (dystrophic) changes in the aged and particularly in the AD brain. No spatial correlation was found between ferritin-positive dystrophic microglia and senile plaques in AD tissues. Analysis of a secondary set of human postmortem brain tissues with a wide range of postmortem intervals (PMI, average 10.94 +/- 5.69 h) showed that the occurrence of microglial dystrophy was independent of PMI and consequently not a product of tissue autolysis. Collectively, these results suggest that microglial involvement in iron storage and metabolism contributes to their degeneration, possibly through increased exposure of the cells to oxidative stress. We conclude that ferritin immunohistochemistry may be a useful method for detecting degenerating microglia in the human brain.
Collapse
Affiliation(s)
- Kryslaine O Lopes
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida 32610-0244, USA
| | | | | |
Collapse
|
36
|
Harry GJ, Kraft AD. Neuroinflammation and microglia: considerations and approaches for neurotoxicity assessment. Expert Opin Drug Metab Toxicol 2008; 4:1265-77. [PMID: 18798697 PMCID: PMC2658618 DOI: 10.1517/17425255.4.10.1265] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The impact of an inflammatory response, as well as interactions between the immune and nervous systems, are rapidly assuming major roles in neurodegenerative disease and injury. However, it is now appreciated that the exact nature of such responses can differ with each type of insult and interaction. More recently, neuroinflammation and the associated cellular response of microglia are being considered for their contribution to neurotoxicity of environmental agents; yet, so far, the inclusion of inflammatory end points into neurotoxicity assessment have relied primarily on relatively limited measures or driven by in vitro models of neurotoxicity. OBJECTIVE To present background information on relevant biological considerations of neuroinflammation and the microglia response demonstrating the complex integrative nature of these biological processes and raising concern with regards to translation of effects demonstrated in vitro to the in vivo situation. Specific points are addressed that would influence the design and interpretation of neuroinflammation with regards to neurotoxicology assessment. CONCLUSION There is a complex and dynamic response in the brain to regulate inflammatory processes and maintain a normal homeostatic level. The classification of such responses as beneficial or detrimental is an oversimplification. Neuroinflammation should be considered as a balanced network of processes in which subtle modifications can shift the cells toward disparate outcomes. The tendency to overinterpret data obtained in an isolated culture system should be discouraged. Rather, the use of cross-disciplinary approaches to evaluate several end points should be incorporated into the assessment of inflammatory contributions to the neurotoxicity of environmental exposures.
Collapse
Affiliation(s)
- Gaylia Jean Harry
- National Institute of Environmental Health Sciences, National Institutes of Health, Neurotoxicology Group, Laboratory of Neurobiology, Department of Health and Human Services, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
37
|
Cipriani S, Bizzoco E, Gianfriddo M, Melani A, Vannucchi M, Pedata F. Adenosine A2A receptor antagonism increases nNOS-immunoreactive neurons in the striatum of Huntington transgenic mice. Exp Neurol 2008; 213:163-70. [DOI: 10.1016/j.expneurol.2008.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 05/16/2008] [Accepted: 05/20/2008] [Indexed: 11/29/2022]
|
38
|
Comparison of ionized calcium-binding adapter molecule 1 immunoreactivity of the hippocampal dentate gyrus and CA1 region in adult and aged dogs. Neurochem Res 2008; 33:1309-15. [PMID: 18270819 DOI: 10.1007/s11064-007-9584-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Accepted: 12/27/2007] [Indexed: 10/22/2022]
Abstract
Similarities between age-related changes in the canine and human brain have resulted in the general acceptance of the canine brain as a model of human brain aging. The hippocampus is essentially required for intact cognitive ability and appears to be particularly vulnerable to the aging process. We observed changes in ionized calcium-binding adapter molecule 1 (Iba-1, a microglial marker) immunoreactivity and protein levels in the hippocampal dentate gyrus and CA1 region of adult (2-3 years) and aged (10-12 years) dogs. We also observed the interferon-gamma (IFN-gamma), a pro-inflammatory cytokine, protein levels in these groups. In the dentate gyrus and CA1 region of the adult dog, Iba-1 immunoreactive microglia were well distributed and their processes were highly ramified. However, in the aged dog, the processes of Iba-1 immunoreactive microglia were hypertrophied in the dentate gyrus. Moreover, Iba-1 protein level in the dentate gyrus in the aged dog was higher than in the adult dog. IFN-gamma expression was increased in the dentate gyrus homogenates of aged dogs than adult dogs. In addition, we found that some neurons were positive to Fluoro-Jade B (a marker for neuronal degeneration) in the dentate polymorphic layer, but not in the hippocampal CA1 region in the aged dog. These results suggest that Iba-1 immunoreactive microglia are hypertrophied in the dentate gyrus in the aged dog.
Collapse
|
39
|
Choi JH, Lee CH, Hwang IK, Won MH, Seong JK, Yoon YS, Lee HS, Lee IS. Age-related changes in ionized calcium-binding adapter molecule 1 immunoreactivity and protein level in the gerbil hippocampal CA1 region. J Vet Med Sci 2008; 69:1131-6. [PMID: 18057827 DOI: 10.1292/jvms.69.1131] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Microglia are evenly distributed throughout the brain parenchyma. They respond rapidly to a variety of alterations in the microenvironment of the brain and act as sensors for pathological events in the brain. In the present study, we investigated the age-dependent changes in the immunoreactivity and protein level of ionized calcium-binding adapter molecule 1 (Iba-1), a microglial marker, in the CA1 region of the gerbil hippocampus. Iba-1 immunoreactive microglia were detected in the hippocampal CA1 region of the postnatal month 1 (PM 1) group. Iba-1 positive microglia were morphologically inactive between the PM 1 and PM 12 stages. Some Iba-1 immunoreactive microglia were present in the active form in the hippocampal CA1 region of the PM 18 and PM 24 groups. The Iba-1 protein levels in hippocampal CA1 homogenates were decreased in the PM 1 through PM 6 groups and increased in an age-dependent manner thereafter. These results suggest that Iba-1 immunoreactive microglia in the active form were detected in the hippocampal CA1 region in the PM 18 and PM 24 groups. This result may be associated with an age-dependent susceptibility to neurodegenerative diseases associated with the hippocampus.
Collapse
Affiliation(s)
- Jung Hoon Choi
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and BK21 Program for Veterinary Science, Seoul National University, seoul 151-742, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Moresco RM, Lavazza T, Belloli S, Lecchi M, Pezzola A, Todde S, Matarrese M, Carpinelli A, Turolla E, Zimarino V, Popoli P, Malgaroli A, Fazio F. Quinolinic acid induced neurodegeneration in the striatum: a combined in vivo and in vitro analysis of receptor changes and microglia activation. Eur J Nucl Med Mol Imaging 2007; 35:704-15. [PMID: 18080815 DOI: 10.1007/s00259-007-0651-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 11/04/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE Huntington's disease (HD) is a progressive neurodegenerative disorder, which is characterised by prominent neuronal cell loss in the basal ganglia with motor and cognitive disturbances. One of the most well-studied pharmacological models of HD is produced by local injection in the rat brain striatum of the excitotoxin quinolinic acid (QA), which produces many of the distinctive features of this human neurodegenerative disorder. Here, we report a detailed analysis, obtained both in vivo and in vitro of this pharmacological model of HD. MATERIALS AND METHODS By combining emission tomography (PET) with autoradiographic and immunocytochemical confocal laser techniques, we quantified in the QA-injected striatum the temporal behavior (from 1 to 60 days from the excitotoxic insult) of neuronal cell density and receptor availability (adenosine A(2A) and dopamine D(2) receptors) together with the degree of microglia activation. RESULTS Both approaches showed a loss of adenosine A(2A) and dopamine D(2) receptors paralleled by an increase of microglial activation. CONCLUSION This combined longitudinal analysis of the disease progression, which suggested an impairment of neurotransmission, neuronal integrity and a reversible activation of brain inflammatory processes, might represent a more quantitative approach to compare the differential effects of treatments in slowing down or reversing HD in rodent models with potential applications to human patients.
Collapse
Affiliation(s)
- R M Moresco
- IBFM-CNR, University of Milan Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Giorgini F, Möller T, Kwan W, Zwilling D, Wacker JL, Hong S, Tsai LCL, Cheah CS, Schwarcz R, Guidetti P, Muchowski PJ. Histone deacetylase inhibition modulates kynurenine pathway activation in yeast, microglia, and mice expressing a mutant huntingtin fragment. J Biol Chem 2007; 283:7390-400. [PMID: 18079112 DOI: 10.1074/jbc.m708192200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The kynurenine pathway of tryptophan degradation is hypothesized to play an important role in Huntington disease, a neurodegenerative disorder caused by a polyglutamine expansion in the protein huntingtin. Neurotoxic metabolites of the kynurenine pathway, generated in microglia and macrophages, are present at increased levels in the brains of patients and mouse models during early stages of disease, but the mechanism by which kynurenine pathway up-regulation occurs in Huntington disease is unknown. Here we report that expression of a mutant huntingtin fragment was sufficient to induce transcription of the kynurenine pathway in yeast and that this induction was abrogated by impairing the activity of the histone deacetylase Rpd3. Moreover, numerous genetic suppressors of mutant huntingtin toxicity that are functionally unrelated converged unexpectedly on the kynurenine pathway, supporting a critical role for the kynurenine pathway in mediating mutant huntingtin toxicity in yeast. Histone deacetylase-dependent regulation of the kynurenine pathway was also observed in a mouse model of Huntington disease, in which treatment with a neuroprotective histone deacetylase inhibitor blocked activation of the kynurenine pathway in microglia expressing a mutant huntingtin fragment in vitro and in vivo. These findings suggest that a mutant huntingtin fragment can perturb transcriptional programs in microglia, and thus implicate these cells as potential modulators of neurodegeneration in Huntington disease that are worthy of further investigation.
Collapse
Affiliation(s)
- Flaviano Giorgini
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Purinergic junctional transmission and propagation of calcium waves in cultured spinal cord microglial networks. Purinergic Signal 2007; 4:47-59. [PMID: 18368533 PMCID: PMC2246000 DOI: 10.1007/s11302-007-9076-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 08/13/2007] [Indexed: 12/27/2022] Open
Abstract
In order to elucidate the mechanisms of purinergic transmission of calcium (Ca(2 + )) waves between microglial cells, we have employed micro-photolithographic methods to form discrete patterns of microglia that allow quantitative measurements of Ca(2 + ) wave propagation. Microglia were confined to lanes 20-100 [Formula: see text] wide and Ca(2 + ) waves propagated from a point of mechanical stimulation, with a diminution in amplitude, for about 120 [Formula: see text]. The number of cells participating in propagation also decreased over this distance. Ca(2 + ) waves could propagate across a cell-free lane from one microglia lane to another if this distance of separation was less than about 60 [Formula: see text], indicating that propagation involved diffusion of a chemical transmitter. This transmitter was identified as ATP since all Ca(2 + ) wave propagation was blocked by the purinoceptor antagonist suramin, which blocks P2Y(2) and P2Y(12) at relatively low concentrations. Antibodies to P2Y(12) showed these at very high density compared with P2Y(2), indicating a role for P2Y(12) receptors. These observations were quantitatively accounted for by a model in which the main determinants are the diffusion of ATP released from a stimulated microglial cell and differences in the dissociation constant of the purinoceptors on the microglial cells.
Collapse
|
43
|
Simmons DA, Casale M, Alcon B, Pham N, Narayan N, Lynch G. Ferritin accumulation in dystrophic microglia is an early event in the development of Huntington's disease. Glia 2007; 55:1074-84. [PMID: 17551926 DOI: 10.1002/glia.20526] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Huntington's Disease (HD) is characterized primarily by neuropathological changes in the striatum, including loss of medium-spiny neurons, nuclear inclusions of the huntingtin protein, gliosis, and abnormally high iron levels. Information about how these conditions interact, or about the temporal order in which they appear, is lacking. This study investigated if, and when, iron-related changes occur in the R6/2 transgenic mouse model of HD and compared the results with those from HD patients. Relative to wild-type mice, R6/2 mice had increased immunostaining for ferritin, an iron storage protein, in the striatum beginning at 2-4 weeks postnatal and in cortex and hippocampus starting at 5-7 weeks. The ferritin staining was found primarily in microglia, and became more pronounced as the mice matured. Ferritin-labeled microglia in R6/2 mice appeared dystrophic in that they had thick, twisted processes with cytoplasmic breaks; some of these cells also contained the mutant huntingtin protein. Brains from HD patients (Vonsattel grades 0-4) also had increased numbers of ferritin-containing microglia, some of which were dystrophic. The cells were positive for Perl's stain, indicating that they contained abnormally high levels of iron. These results provide the first evidence that perturbations to iron metabolism in HD are predominately associated with microglia and occur early enough to be important contributors to HD progression.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697-4292, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Priller J, Prinz M, Heikenwalder M, Zeller N, Schwarz P, Heppner FL, Aguzzi A. Early and rapid engraftment of bone marrow-derived microglia in scrapie. J Neurosci 2006; 26:11753-62. [PMID: 17093096 PMCID: PMC6674798 DOI: 10.1523/jneurosci.2275-06.2006] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prion neuroinvasion is accompanied by maximal activation of microglia, the significance of which for pathogenesis is unknown. Here, we used bone marrow (BM) cells expressing GFP (green fluorescent protein) to study the turnover of microglia in mouse scrapie. We found that >or=50% of all brain microglia were replaced by BM-derived cells before clinical disease onset. In terminally sick mice, microglia density increased threefold to fourfold. Hence BM-derived microglia rapidly and efficaciously colonize the brain in scrapie. Whereas reconstitution of wild-type mice with prion protein-deficient (Prnp(o/o)) BM did not alter scrapie pathogenesis, Prnp(o/o) mice transplanted with wild-type BM cells were resistant to peripherally administered prions despite high levels of infectivity in the spleen. Cerebellar homogenates from prion-inoculated Prnp(o/o) mice reconstituted with >10% of wild-type microglia failed to infect transgenic mice overexpressing the cellular prion protein. Hence, in contrast to previous reports, microglia are not competent for efficient prion transport and replication in vivo.
Collapse
Affiliation(s)
- Josef Priller
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
- Departments of Psychiatry and Experimental Neurology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany, and
| | - Marco Prinz
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
- Institute of Neuropathology, University of Göttingen, 37075 Göttingen, Germany
| | - Mathias Heikenwalder
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Nicolas Zeller
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Frank L. Heppner
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, Department of Pathology, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
45
|
Batista CMC, Kippin TE, Willaime-Morawek S, Shimabukuro MK, Akamatsu W, van der Kooy D. A progressive and cell non-autonomous increase in striatal neural stem cells in the Huntington's disease R6/2 mouse. J Neurosci 2006; 26:10452-60. [PMID: 17035529 PMCID: PMC6674685 DOI: 10.1523/jneurosci.2850-06.2006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neural stem and progenitor cells are located in the subependyma of the adult forebrain. An increase in adult subependymal cell proliferation is reported after various kinds of brain injury. We demonstrate an expansion of neural precursor cells in the postnatal subependyma in a murine genetic disease model of Huntington's disease (HD), the R6/2 mouse. We used the in vitro neurosphere assay as an index of the number of neural stem cells in vivo and to assess proliferation kinetics in vitro and in vivo bromodeoxyuridine labeling to assess the progenitor cell population and their fates. Disease progression in this model leads to an increase in the numbers of neural stem cells in the adult striatal subependyma. This increase is produced cell non-autonomously by events in the R6/2 brains as the mice become increasingly symptomatic. Once the neural stem cell increase is induced in vivo, it is maintained during in vitro passaging of neural stem cells, but the neural stem cell increase is not reproduced during in vitro passaging of neural stem cells from presymptomatic R6/2 mice. In addition, we show that some of the R6/2 neural progenitor cells show a change from their normal migration destiny toward the olfactory bulb. Instead, some of these cells migrate into the striatum, one of the main affected areas in HD. Our findings demonstrate that HD damage recruits precursor cells in two ways: expansion of neural stem cells and altered migration of progenitor cells.
Collapse
Affiliation(s)
- Claudia M. C. Batista
- Neurobiology Research Group, Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada M5S 3E1
- Departmento de Histologia e Embriologia, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, 21941-590 Rio de Janeiro, Brazil, and
| | - Tod E. Kippin
- Neurobiology Research Group, Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada M5S 3E1
- Department of Psychology, University of California, Santa Barbara, Santa Barbara, California 93106-9660
| | - Sandrine Willaime-Morawek
- Neurobiology Research Group, Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Marília Kimie Shimabukuro
- Departmento de Histologia e Embriologia, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, 21941-590 Rio de Janeiro, Brazil, and
| | - Wado Akamatsu
- Neurobiology Research Group, Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| | - Derek van der Kooy
- Neurobiology Research Group, Department of Medical Genetics and Microbiology, University of Toronto, Toronto, Ontario, Canada M5S 3E1
| |
Collapse
|
46
|
Abstract
The aging brain is characterized by a demonstrable decrease in weight and volume, particularly after the age of 50. This atrophy, which affects both grey and white matter, is presumed to result from a loss of neurons and myelinated axons. Glial cells, on the other hand, appear to increase in the aging brain, which exhibits greater immunoreactivity with both astrocytic and microglial markers. This review is focused on the morphologic and phenotypic changes that occur in microglial cells with normal aging. Although there is a consistent aging-related upregulation of microglial activation markers in experimental animals and humans that could be interpreted as aging-related neuroinflammation, it is generally difficult to show a direct correlation between ostensible microglial activation and neurodegeneration. This raises questions about whether aging-related microglial activation indeed represents reactive gliosis in the conventional sense. As an alternative, we discuss the possibility that structural and phenotypic changes that occur in microglia are a direct reflection of the aging process on microglia. Thus, microglia cells themselves may be subject to cellular senescence in the sense that they no longer function efficiently. The concept of microglial senescence offers a novel perspective on aging-related neurodegeneration, namely that neurodegeneration could also occur secondary to microglial degeneration.
Collapse
Affiliation(s)
- Jessica R Conde
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, 32610, USA
| | | |
Collapse
|
47
|
Phillips W, Morton AJ, Barker RA. Abnormalities of neurogenesis in the R6/2 mouse model of Huntington's disease are attributable to the in vivo microenvironment. J Neurosci 2006; 25:11564-76. [PMID: 16354914 PMCID: PMC6726042 DOI: 10.1523/jneurosci.3796-05.2005] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative condition characterized by movement disorders, psychiatric disturbance, and cognitive decline. There are no treatments to halt or reverse the disease. Mammalian neurogenesis persists into adulthood in the subventricular zone (SVZ) and dentate gyrus (DG) of the hippocampus. In 2001, our laboratory published the hypothesis that neurogenesis is impaired in neurodegenerative diseases and that this may contribute to disease progression. Since then, it has been shown that neurogenesis is reduced in the DG of transgenic HD mice but increased in the SVZ of HD patients. We sought to characterize neurogenesis further. We found that, in the DG of the transgenic R6/2 mouse model of HD, newborn cell proliferation and morphology, but not differentiation or survival, was compromised. In R6/2 mice, neurogenesis failed to upregulate in the DG in response to seizures. Basal SVZ neurogenesis was similar between R6/2 mice and their wild-type littermates. There was no difference in the in vitro growth of adult neural precursor cells (NPCs) between genotypes. These results suggest that abnormal neurogenesis in the R6/2 mouse is not attributable to an intrinsic impairment of the NPC itself but is attributable to the environment in which the cell is located.
Collapse
Affiliation(s)
- Wendy Phillips
- Cambridge Centre for Brain Repair, Cambridge CB2 2PY, United Kingdom.
| | | | | |
Collapse
|
48
|
Crocker SF, Costain WJ, Robertson HA. DNA microarray analysis of striatal gene expression in symptomatic transgenic Huntington's mice (R6/2) reveals neuroinflammation and insulin associations. Brain Res 2006; 1088:176-86. [PMID: 16626669 DOI: 10.1016/j.brainres.2006.02.102] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 02/03/2006] [Accepted: 02/26/2006] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is an inherited, progressive neurodegenerative disorder caused by CAG repeat expansion in the gene that codes for the protein huntingtin. The underlying neuropathological events leading to the selectivity of striatal neuronal loss are unknown. However, the huntingtin mutation interferes at several levels of normal cell function. The complexity of this disease makes microarray analysis an appealing technique to begin the identification of common pathways that may contribute to the pathology. In this study, striatal tissue was extracted for gene expression profiling from wild-type and symptomatic transgenic Huntington mice (R6/2) expressing part of the human Huntington's disease gene. We interrogated a 15 K high-density mouse EST array not previously used for HD and identified 170 significantly differentially expressed ESTs in symptomatic R6/2 mice. Of the 80 genes with known function, 9 genes had previously been identified as altered in HD. 71 known genes were associated with HD for the first time. The data obtained from this study confirm and extend previous observations using DNA microarray techniques on genetic models for HD, revealing novel changes in expression in a number of genes not previously associated with HD. Further bioinformatic analysis, using software to construct biological association maps, focused attention on proteins such as insulin and TH1-mediated cytokines, suggesting that they may be important regulators of affected genes. These results may provide insight into the regulation and interaction of genes that contribute to adaptive and pathological processes involved in HD.
Collapse
Affiliation(s)
- Susan F Crocker
- Brain Repair Centre, Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | | | | |
Collapse
|
49
|
Glass R, Synowitz M, Kronenberg G, Walzlein JH, Markovic DS, Wang LP, Gast D, Kiwit J, Kempermann G, Kettenmann H. Glioblastoma-induced attraction of endogenous neural precursor cells is associated with improved survival. J Neurosci 2006; 25:2637-46. [PMID: 15758174 PMCID: PMC6725181 DOI: 10.1523/jneurosci.5118-04.2005] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neural precursor cells contribute to adult neurogenesis and to limited attempts of brain repair after injury. Here we report that in a murine experimental glioblastoma model, endogenous neural precursors migrate from the subventricular zone toward the tumor and surround it. The association of endogenous precursors with syngenic tumor grafts was observed, after injecting red fluorescent protein-labeled G261 cells into the caudate-putamen of transgenic mice, which express green fluorescent protein under a promoter for nestin (nestin-GFP). Fourteen days after inoculation, the nestin-GFP cells surrounded the tumors in several cell layers and expressed markers of early noncommitted and committed precursors. Nestin-GFP cells were further identified by a characteristic membrane current pattern as recorded in acute brain slices. 5-bromo-2-deoxyuridine labeling and dye tracing experiments revealed that the tumor-associated precursors originated from the subventricular zone. Moreover, in cultured explants from the subventricular zone, the neural precursors showed extensive tropism for glioblastomas. Tumor-induced endogenous precursor cell accumulation decreased with age of the recipient; this correlated with increased tumor size and shorter survival times in aged mice. Coinjection of glioblastoma cells with neural precursors improved the survival time of old mice to a level similar to that in young mice. Coculture experiments showed that neural precursors suppressed the rapid increase in tumor cell number, which is characteristic of glioblastoma, and induced glioblastoma cell apoptosis. Our results indicate that tumor cells attract endogenous precursor cells; the presence of precursor cells is antitumorigenic; and this cellular interaction decreases with aging.
Collapse
Affiliation(s)
- Rainer Glass
- Cellular Neuroscience Group, Max Delbrück Center for Molecular Medicine, 13092 Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Schwartz M, Butovsky O, Brück W, Hanisch UK. Microglial phenotype: is the commitment reversible? Trends Neurosci 2006; 29:68-74. [PMID: 16406093 DOI: 10.1016/j.tins.2005.12.005] [Citation(s) in RCA: 328] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2005] [Revised: 10/26/2005] [Accepted: 12/15/2005] [Indexed: 11/22/2022]
Abstract
Microglia, the standby cells for immune defense in the CNS, have a reputation for exacerbating the neural damage that occurs in neurodegenerative diseases. However, research over the past few years has established that microglia do not constitute a single, uniform cell population, but rather comprise a family of cells with diverse phenotypes--some that are beneficial and others that the CNS can barely tolerate and that are therefore destructive. This finding raised several questions. What instructs microglia to acquire a particular phenotype, and how do these phenotypes differ? How committed are microglia to a specific phenotype? Can destructive microglia become protective, and can protective microglia retain their beneficial phenotype even when they encounter a destructive environment? Here, we address these questions, and the background of research that elicited them.
Collapse
Affiliation(s)
- Michal Schwartz
- The Weizmann Institute of Science, POB 26, Rehovot, 76100, Israel.
| | | | | | | |
Collapse
|