1
|
Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol 2024; 147:66. [PMID: 38568475 PMCID: PMC10991006 DOI: 10.1007/s00401-024-02712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/03/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Reactive astrogliosis accompanies the two neuropathological hallmarks of Alzheimer's disease (AD)-Aβ plaques and neurofibrillary tangles-and parallels neurodegeneration in AD and AD-related dementias (ADRD). Thus, there is growing interest in developing imaging and fluid biomarkers of reactive astrogliosis for AD/ADRD diagnosis and prognostication. Monoamine oxidase-B (MAO-B) is emerging as a target for PET imaging radiotracers of reactive astrogliosis. However, a thorough characterization of MAO-B expression in postmortem control and AD/ADRD brains is lacking. We sought to: (1) identify the primary cell type(s) expressing MAO-B in control and AD brains; (2) quantify MAO-B immunoreactivity in multiple brain regions of control and AD donors as a proxy for PET radiotracer uptake; (3) correlate MAO-B level with local AD neuropathological changes, reactive glia, and cortical atrophy; (4) determine whether the MAOB rs1799836 SNP genotype impacts MAO-B expression level; (5) compare MAO-B immunoreactivity across AD/ADRD, including Lewy body diseases (LBD) and frontotemporal lobar degenerations with tau (FTLD-Tau) and TDP-43 (FTLD-TDP). We found that MAO-B is mainly expressed by subpial and perivascular cortical astrocytes as well as by fibrous white matter astrocytes in control brains, whereas in AD brains, MAO-B is significantly upregulated by both cortical reactive astrocytes and white matter astrocytes across temporal, frontal, and occipital lobes. By contrast, MAO-B expression level was unchanged and lowest in cerebellum. Cortical MAO-B expression was independently associated with cortical atrophy and local measures of reactive astrocytes and microglia, and significantly increased in reactive astrocytes surrounding Thioflavin-S+ dense-core Aβ plaques. MAO-B expression was not affected by the MAOB rs1799836 SNP genotype. MAO-B expression was also significantly increased in the frontal cortex and white matter of donors with corticobasal degeneration, Pick's disease, and FTLD-TDP, but not in LBD or progressive supranuclear palsy. These findings support ongoing efforts to develop MAO-B-based PET radiotracers to image reactive astrogliosis in AD/ADRD.
Collapse
Affiliation(s)
- Methasit Jaisa-Aad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Muñoz-Castro
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Molly A Healey
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
2
|
Kumar A, Fontana IC, Nordberg A. Reactive astrogliosis: A friend or foe in the pathogenesis of Alzheimer's disease. J Neurochem 2023; 164:309-324. [PMID: 34931315 DOI: 10.1111/jnc.15565] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/28/2022]
Abstract
Astrocytes are highly efficient homeostatic glial cells playing a crucial role in optimal brain functioning and homeostasis. Astrocytes respond to changes in brain homoeostasis following central nervous system (CNS) injury/diseased state by a specific defence mechanism called reactive astrogliosis. Recent studies have implicated and placed reactive astrogliosis in the centre of pathophysiology of Alzheimer's disease (AD) and other neurodegenerative disorders. The AD biomarker field is evolving rapidly with new findings providing strong evidence which supports the notion that a reactive astrogliosis is an early event in the time course of AD progression which may precede other pathological hallmarks of AD. Clinical/translational in vivo PET and in vitro postmortem brain imaging studies demonstrated 'a first and second wave' of reactive astrogliosis in AD with distinct close-loop relationships with other pathological biomarkers at different stages of the disease. At the end stages, reactive astrocytes are found to be associated, or in proximity, with amyloid plaque and tau pathological deposits in postmortem AD brains. Several new PET-tracers, which are being in pipeline and validated at a very fast pace for mapping and visualising reactive astrogliosis in the brain, will provide further invaluable mechanistic insights into AD and other non-AD dementia pathologies. The complementary roles of microglia and astrocyte activation in AD progression, along with the clinical value of new fluid astrocytes biomarkers in the context of existing biomarkers, are the latest avenue that needs further exploration.
Collapse
Affiliation(s)
- Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Igor C Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Nwaubani P, Cercignani M, Colasanti A. In vivo quantitative imaging of hippocampal inflammation in autoimmune neuroinflammatory conditions: a systematic review. Clin Exp Immunol 2022; 210:24-38. [PMID: 35802780 PMCID: PMC9585553 DOI: 10.1093/cei/uxac058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 01/25/2023] Open
Abstract
The hippocampus is a morphologically complex region of the brain limbic system centrally involved in important cognitive, affective, and behavioural regulatory roles. It has exquisite vulnerability to neuroinflammatory processes, with some of its subregions found to be specific sites of neuroinflammatory pathology in ex-vivo studies. Optimizing neuroimaging correlates of hippocampal neuroinflammation would enable the direct study of functional consequences of hippocampal neuroinflammatory pathology, as well as the definition of therapeutic end-points for treatments targeting neuroinflammation, and their related affective or cognitive sequelae. However, in vivo traditional imaging of the hippocampus and its subregions is fraught with difficulties, due to methodological challenges deriving from its unique anatomical characteristics. The main objective of this review is to provide a current update on the characterization of quantitative neuroimaging correlates of hippocampal neuroinflammation by focusing on three prototypical autoimmune neuro-inflammatory conditions [multiple sclerosis (MS), systemic lupus erythematosus (SLE), and autoimmune encephalitis (AE)]. We focused on studies employing TSPO-targeting positron emission tomography (PET), quantitative magnetic resonance imaging (MRI), and spectroscopy techniques assumed to be sensitive to neuroinflammatory tissue changes. We found 18 eligible studies (14, 2, and 2 studies in MS, AE, and SLE, respectively). Across conditions, the largest effect was seen in TSPO PET and diffusion-weighted MRI studies. No study examined neuroinflammation-related changes at the hippocampal subfield level. Overall, results were largely inconsistent due to heterogeneous imaging methods, small sample sizes, and different population studies. We discuss how these data could inform future study design and conclude by suggesting further methodological directions aimed at improving the precision and sensitivity of neuroimaging techniques to characterize hippocampal neuroinflammatory pathology in the human brain.
Collapse
Affiliation(s)
- P Nwaubani
- Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, UK
| | - M Cercignani
- Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK
| | - A Colasanti
- Correspondence: Alessandro Colasanti, Department of Clinical Neuroscience and Neuroimaging, Brighton and Sussex Medical School, University of Sussex, Trafford Centre for Medical Research, University of Sussex, Falmer, Brighton, BN1 4RY, UK.
| |
Collapse
|
4
|
Wang Y, Sadaghiani MS, Tian F, Fitzgerald KC, Solnes L, Newsome SD. Brain and Muscle Metabolic Changes by FDG-PET in Stiff Person Syndrome Spectrum Disorders. Front Neurol 2021; 12:692240. [PMID: 34603180 PMCID: PMC8484315 DOI: 10.3389/fneur.2021.692240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/30/2021] [Indexed: 11/30/2022] Open
Abstract
Objective: To report clinical characteristics and fluorodeoxyglucose positron emission tomography (FDG-PET) findings in the brain and muscles of individuals with stiff person syndrome (SPS) spectrum disorders (SPSSDs). Methods: Retrospective cohort study from 1997 to 2018 at Johns Hopkins Hospital identified 170 individuals with SPS or cerebellar ataxia (CA) associated with anti-glutamic acid decarboxylase (anti-GAD)-65 antibodies. Fifty-one underwent FDG-PET, with 50 involving the body and 30 with dedicated brain acquisition. The clinical and immunological profiles were extracted via medical record review. The brain scans were analyzed quantitatively using the NeuroQ software, with comparison with an averaged normal database. The body scans were reviewed qualitatively by a blinded nuclear medicine radiologist. Results: Mean age of symptom onset was 41.5 years (range 12–75 years). Majority were female (68%) and White (64%). Of the patients, 82% had SPS (majority being classic phenotype), and 18% had CA. Three had a paraneoplastic process. Forty-seven had serum anti-GAD, two with anti-amphiphysin, and one with anti-glycine receptor antibodies. Brain metabolic abnormalities were seen in both SPS and CA, with significant differences between the groups noted in the right superior frontal cortex, right sensorimotor cortex, left inferior parietal cortex, bilateral thalami, vermis, and left cerebellum. Of the patients, 62% demonstrated muscle hypermetabolism, most commonly bilateral, involving the upper extremities or axial muscles. Neither brain nor muscle metabolism was correlated with functional outcomes nor treatments. Conclusions: Metabolic changes as seen by FDG-PET are present in the brain and muscle in many individuals with SPSSD. Future studies are needed to assess whether FDG-PET can help aid in the diagnosis and/or monitoring of individuals with SPSSD.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mohammad S Sadaghiani
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Fan Tian
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lilja Solnes
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Scott D Newsome
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Verkerke M, Hol EM, Middeldorp J. Physiological and Pathological Ageing of Astrocytes in the Human Brain. Neurochem Res 2021; 46:2662-2675. [PMID: 33559106 PMCID: PMC8437874 DOI: 10.1007/s11064-021-03256-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
Ageing is the greatest risk factor for dementia, although physiological ageing by itself does not lead to cognitive decline. In addition to ageing, APOE ε4 is genetically the strongest risk factor for Alzheimer's disease and is highly expressed in astrocytes. There are indications that human astrocytes change with age and upon expression of APOE4. As these glial cells maintain water and ion homeostasis in the brain and regulate neuronal transmission, it is likely that age- and APOE4-related changes in astrocytes have a major impact on brain functioning and play a role in age-related diseases. In this review, we will discuss the molecular and morphological changes of human astrocytes in ageing and the contribution of APOE4. We conclude this review with a discussion on technical issues, innovations, and future perspectives on how to gain more knowledge on astrocytes in the human ageing brain.
Collapse
Affiliation(s)
- Marloes Verkerke
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Immunobiology, Biomedical Primate Research Centre (BPRC), P.O. Box 3306, 2280 GH, Rijswijk, The Netherlands
| |
Collapse
|
6
|
Prabhakaran J, Molotkov A, Mintz A, Mann JJ. Progress in PET Imaging of Neuroinflammation Targeting COX-2 Enzyme. Molecules 2021; 26:molecules26113208. [PMID: 34071951 PMCID: PMC8198977 DOI: 10.3390/molecules26113208] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 01/01/2023] Open
Abstract
Neuroinflammation and cyclooxygenase-2 (COX-2) upregulation are associated with the pathogenesis of degenerative brain diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), epilepsy, and a response to traumatic brain injury (TBI) or stroke. COX-2 is also induced in acute pain, depression, schizophrenia, various cancers, arthritis and in acute allograft rejection. Positron emission tomography (PET) imaging allows for the direct measurement of in vivo COX-2 upregulation and thereby enables disease staging, therapy evaluation and aid quantifying target occupancy of novel nonsteroidal anti-inflammatory drugs or NSAIDs. Thus far, no clinically useful radioligand is established for monitoring COX-2 induction in brain diseases due to the delay in identifying qualified COX-2-selective inhibitors entering the brain. This review examines radiolabeled COX-2 inhibitors reported in the past decade and identifies the most promising radioligands for development as clinically useful PET radioligands. Among the radioligands reported so far, the three tracers that show potential for clinical translation are, [11CTMI], [11C]MC1 and [18F]MTP. These radioligands demonstrated BBB permeablity and in vivo binding to constitutive COX-2 in the brain or induced COX-2 during neuroinflammation.
Collapse
Affiliation(s)
- Jaya Prabhakaran
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA;
- Correspondence:
| | - Andrei Molotkov
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA; (A.M.); (A.M.)
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY 10032, USA; (A.M.); (A.M.)
- Area Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA
| | - J. John Mann
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA;
- Area Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
7
|
Chatterjee P, Pedrini S, Stoops E, Goozee K, Villemagne VL, Asih PR, Verberk IMW, Dave P, Taddei K, Sohrabi HR, Zetterberg H, Blennow K, Teunissen CE, Vanderstichele HM, Martins RN. Plasma glial fibrillary acidic protein is elevated in cognitively normal older adults at risk of Alzheimer's disease. Transl Psychiatry 2021; 11:27. [PMID: 33431793 PMCID: PMC7801513 DOI: 10.1038/s41398-020-01137-1] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/26/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP), an astrocytic cytoskeletal protein, can be measured in blood samples, and has been associated with Alzheimer's disease (AD). However, plasma GFAP has not been investigated in cognitively normal older adults at risk of AD, based on brain amyloid-β (Aβ) load. Cross-sectional analyses were carried out for plasma GFAP and plasma Aβ1-42/Aβ1-40 ratio, a blood-based marker associated with brain Aβ load, in participants (65-90 years) categorised into low (Aβ-, n = 63) and high (Aβ+, n = 33) brain Aβ load groups via Aβ positron emission tomography. Plasma GFAP, Aβ1-42, and Aβ1-40 were measured using the Single molecule array (Simoa) platform. Plasma GFAP levels were significantly higher (p < 0.00001), and plasma Aβ1-42/Aβ1-40 ratios were significantly lower (p < 0.005), in Aβ+ participants compared to Aβ- participants, adjusted for covariates age, sex, and apolipoprotein E-ε4 carriage. A receiver operating characteristic curve based on a logistic regression of the same covariates, the base model, distinguished Aβ+ from Aβ- (area under the curve, AUC = 0.78), but was outperformed when plasma GFAP was added to the base model (AUC = 0.91) and further improved with plasma Aβ1-42/Aβ1-40 ratio (AUC = 0.92). The current findings demonstrate that plasma GFAP levels are elevated in cognitively normal older adults at risk of AD. These observations suggest that astrocytic damage or activation begins from the pre-symptomatic stage of AD and is associated with brain Aβ load. Observations from the present study highlight the potential of plasma GFAP to contribute to a diagnostic blood biomarker panel (along with plasma Aβ1-42/Aβ1-40 ratios) for cognitively normal older adults at risk of AD.
Collapse
Affiliation(s)
- Pratishtha Chatterjee
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia
| | - Steve Pedrini
- grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia
| | | | - Kathryn Goozee
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia ,grid.489025.2KaRa Institute of Neurological Diseases, Macquarie Park, NSW Australia ,Anglicare, Castle Hill Sydney, NSW Australia ,grid.1012.20000 0004 1936 7910School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA Australia ,The Cooperative Research Centre for Mental Health, Carlton South, Australia
| | - Victor L. Villemagne
- grid.410678.cDepartment of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC Australia
| | - Prita R. Asih
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia
| | - Inge M. W. Verberk
- grid.484519.5Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Preeti Dave
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia ,Anglicare, Castle Hill Sydney, NSW Australia
| | - Kevin Taddei
- grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia ,grid.429545.b0000 0004 5905 2729Australian Alzheimer’s Research Foundation, Nedlands, WA Australia
| | - Hamid R. Sohrabi
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia ,grid.429545.b0000 0004 5905 2729Australian Alzheimer’s Research Foundation, Nedlands, WA Australia ,grid.1025.60000 0004 0436 6763Centre for Healthy Ageing, School of Psychology and Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Murdoch, WA Australia
| | - Henrik Zetterberg
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden ,grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden ,grid.83440.3b0000000121901201Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom ,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- grid.8761.80000 0000 9919 9582Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden ,grid.1649.a000000009445082XClinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Charlotte E. Teunissen
- grid.484519.5Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | | | - Ralph N. Martins
- grid.1004.50000 0001 2158 5405Department of Biomedical Sciences, Macquarie University, North Ryde, NSW Australia ,grid.1038.a0000 0004 0389 4302School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA Australia ,grid.489025.2KaRa Institute of Neurological Diseases, Macquarie Park, NSW Australia ,grid.1012.20000 0004 1936 7910School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, WA Australia ,The Cooperative Research Centre for Mental Health, Carlton South, Australia ,grid.429545.b0000 0004 5905 2729Australian Alzheimer’s Research Foundation, Nedlands, WA Australia
| |
Collapse
|
8
|
Hashioka S, Wu Z, Klegeris A. Glia-Driven Neuroinflammation and Systemic Inflammation in Alzheimer's Disease. Curr Neuropharmacol 2021; 19:908-924. [PMID: 33176652 PMCID: PMC8686312 DOI: 10.2174/1570159x18666201111104509] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/28/2020] [Accepted: 11/06/2020] [Indexed: 11/29/2022] Open
Abstract
The neuroinflammatory hypothesis of Alzheimer's disease (AD) was proposed more than 30 years ago. The involvement of the two main types of glial cells microglia and astrocytes, in neuroinflammation, was suggested early on. In this review, we highlight that the exact contributions of reactive glia to AD pathogenesis remain difficult to define, likely due to the heterogeneity of glia populations and alterations in their activation states through the stages of AD progression. In the case of microglia, it is becoming apparent that both beneficially and adversely activated cell populations can be identified at various stages of AD, which could be selectively targeted to either limit their damaging actions or enhance beneficial functions. In the case of astrocytes, less information is available about potential subpopulations of reactive cells; it also remains elusive whether astrocytes contribute to the neuropathology of AD by mainly gaining neurotoxic functions or losing their ability to support neurons due to astrocyte damage. We identify L-type calcium channel blocker, nimodipine, as a candidate drug for AD, which potentially targets both astrocytes and microglia. It has already shown consistent beneficial effects in basic experimental and clinical studies. We also highlight the recent evidence linking peripheral inflammation and neuroinflammation. Several chronic systemic inflammatory diseases, such as obesity, type 2 diabetes mellitus, and periodontitis, can cause immune priming or adverse activation of glia, thus exacerbating neuroinflammation and increasing risk or facilitating the progression of AD. Therefore, reducing peripheral inflammation is a potentially effective strategy for lowering AD prevalence.
Collapse
Affiliation(s)
- Sadayuki Hashioka
- Address correspondence to these authors at the Department of Psychiatry, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan;, E-mail: and Department of Biology, Faculty of Science, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada; E-mail:
| | | | - Andis Klegeris
- Address correspondence to these authors at the Department of Psychiatry, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan;, E-mail: and Department of Biology, Faculty of Science, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada; E-mail:
| |
Collapse
|
9
|
Abstract
Pathological accumulated misfolded tau underlies various neurodegenerative diseases and associated clinical syndromes. To diagnose those diseases reliable before death or even at early stages, many different tau-specific radiotracers have been developed in the last decade to be used with positron-emission-tomography. In contrast to amyloid-β imaging, different isoforms of tau exist further complicating radiotracer development. First-generation radiotracers like [11C]PBB3, [18F]AV1451 and [18F]THK5351 have been extensively investigated in vitro and in vivo. In Alzheimer's disease (AD), high specific binding could be detected, and evidence of clinical applicability recently led to clinical approval of [18F]flortaucipir ([18F]AV1451) by the FDA. Nevertheless, absent or minor binding to non-AD tau isoforms and high off-target binding to non-tau brain structures limit the diagnostic applicability especially in non-AD tauopathies demanding further tracer development. In vitro assays and autoradiography results of next-generation radiotracers [18F]MK-6240, [18F]RO-948, [18F]PM-PBB3, [18F]GTP-1 and [18F]PI-2620 clearly indicate less off-target binding and high specific binding to tau neurofibrils. First in human studies have been conducted with promising results for all tracers in AD patients, and also some positive experience in non-AD tauopathies. Overall, larger scaled autoradiography and human studies are needed to further evaluate the most promising candidates and support future clinical approval.
Collapse
Affiliation(s)
- Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
10
|
Cheng JJ, Guo Q, Wu XG, Ma S, Gao Y, Ya-Zhen S. Scutellaria barbata flavonoids improve the composited Aβ-induced abnormal changes of glial cells in rats' brain. Comb Chem High Throughput Screen 2020; 25:64-76. [PMID: 33297910 DOI: 10.2174/1386207323666201209092358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/21/2020] [Accepted: 11/15/2020] [Indexed: 11/22/2022]
Abstract
AIM It has been reported that glial cells are involved in Alzheimer's disease (AD). According to our previous research, Scutellaria barbata flavonoids (SBFs) can protect the neuronal disorder and memory impairment for AD-like rats, while the effect of SBFs on the glial cells disorder in AD-like rats has been less well studied. The effects of SBFs on astrocytes(ASs), microglial cells (MGs) and oligodendrocytes (Ols), as well as heat shock proteins 70 (Hsp70) and apolipoprotein E (ApoE) were investigated in the present study. METHODS The successful model rats, screened by Morris water maze, were daily orally administrated with 35, 70 and 140 mg/kg SBFs for 36 d. The numbers of brain's astrocytes (ASs), microglial cells (MGs) and oligodendrocytes (Ols) were examined by immunohistochemistry. The cortical glial fibrillary acidic protein (GFAP), leukocyte common antigen (LCA) (CD45), Claudin 11 and heat shock proteins 70 (Hsp70) protein expression were assayed by Western blotting, and apolipoprotein E (ApoE) mRNA expression was analyzed by real-time quantitative polymerase chain reaction (qPCR). RESULTS Compared with the sham-operated group, the numbers of ASs and MGs in the brain were significantly increased in the model group (P<0.05, P<0.01), and accompanied with increases of GFAP, CD45 and Hsp70 protein and ApoE mRNA expression (P<0.05, P<0.01). Both Ols number and Claudin 11 protein expression decreased in the brain in the model group (P<0.05, P<0.01). However, the above abnormal changes induced by composited Aβ were differently reversed by treatment of SBFs at three doses of 35, 70 and 140 mg/kg (P<0.05, P<0.01). CONCLUSIONS SBFs can dramatically improve the abnormal changes of glial cells in rats' brain induced by composited Aβ, which may be a helpful treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian-Jun Cheng
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Qing Guo
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011. China
| | - Xiao-Guang Wu
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Shuai Ma
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Yang Gao
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| | - Shang Ya-Zhen
- Institute of Traditional Chinese Medicine, Chengde Medical College, Hebei Province Key Research Office of Traditional Chinese Medicine Against Dementia, Hebei Province Key Laboratory of Traditional Chinese Medicine Research and Development, Chengde, Hebei 067000. China
| |
Collapse
|
11
|
Valenza M, Facchinetti R, Steardo L, Scuderi C. Altered Waste Disposal System in Aging and Alzheimer's Disease: Focus on Astrocytic Aquaporin-4. Front Pharmacol 2020; 10:1656. [PMID: 32063858 PMCID: PMC7000422 DOI: 10.3389/fphar.2019.01656] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022] Open
Abstract
Among the diverse cell types included in the general population named glia, astrocytes emerge as being the focus of a growing body of research aimed at characterizing their heterogeneous and complex functions. Alterations of both their morphology and activities have been linked to a variety of neurological diseases. One crucial physiological need satisfied by astrocytes is the cleansing of the cerebral tissue from waste molecules. Several data demonstrate that aquaporin-4 (AQP-4), a protein expressed by astrocytes, is crucially important for facilitating the removal of waste products from the brain. Aquaporins are water channels found in all district of the human organism and the most abundant isoform in the brain is AQP-4. This protein is involved in a myriad of astrocytic activities, including calcium signal transduction, potassium buffering, synaptic plasticity, astrocyte migration, glial scar formation and neuroinflammation. The highest density of AQP-4 is found at the astrocytic domains closest to blood vessels, the endfeet that envelop brain vessels, with low to zero expression in other astrocytic membrane regions. Increased AQP-4 expression and loss of polarization have recently been documented in altered physiological conditions. Here we review the latest findings related to aging and Alzheimer’s disease (AD) on this topic, as well as the available knowledge on pharmacological tools to target AQP-4.
Collapse
Affiliation(s)
- Marta Valenza
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy.,Epitech Group SpA, Saccolongo, Italy
| | - Roberta Facchinetti
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Luca Steardo
- Università Telematica Giustino Fortunato, Benevento, Italy
| | - Caterina Scuderi
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
12
|
Schönecker S, Brendel M, Palleis C, Beyer L, Höglinger GU, Schuh E, Rauchmann BS, Sauerbeck J, Rohrer G, Sonnenfeld S, Furukawa K, Ishiki A, Okamura N, Bartenstein P, Dieterich M, Bötzel K, Danek A, Rominger A, Levin J. PET Imaging of Astrogliosis and Tau Facilitates Diagnosis of Parkinsonian Syndromes. Front Aging Neurosci 2019; 11:249. [PMID: 31572166 PMCID: PMC6749151 DOI: 10.3389/fnagi.2019.00249] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/22/2019] [Indexed: 11/13/2022] Open
Abstract
Neurodegenerative parkinsonian syndromes comprise a number of disorders that are characterized by similar clinical features but are separated on the basis of different pathologies, i.e., aggregates of α-synuclein or tau protein. Due to the overlap of signs and symptoms a precise differentiation is often difficult, especially early in the disease course. Enormous efforts have been taken to develop tau-selective PET imaging agents, but strong off-target binding to monoamine oxidase B (MAO-B) has been observed across first generation ligands. Nonetheless, astrogliosis-related MAO-B elevation is a common histopathological known feature of all parkinsonian syndromes and might be itself an interesting imaging target. Therefore, this study aimed to investigate the performance of [18F]-THK5351, a combined MAO-B and tau tracer for differential diagnosis of parkinsonian syndromes. [18F]-THK5351 PET was performed in 34 patients: six with Parkinson's disease (PD), nine with multiple system atrophy with predominant parkinsonism (MSA-P), six with MSA with predominant cerebellar ataxia (MSA-C), and 13 with progressive supranuclear palsy (PSP) Richardson's syndrome. Volume-of-interest-based quantification of standardized-uptake-values was conducted in different parkinsonian syndrome-related target regions. PET results were subjected to multinomial logistic regression to create a prediction model discriminating among groups. Furthermore, we correlated tracer uptake with clinical findings. Elevated [18F]-THK5351 uptake in midbrain and diencephalon differentiated PSP patients from PD and MSA-C. MSA-C patients were distinguishable by high tracer uptake in the pons and cerebellar deep white matter when compared to PSP and PD patients, whereas MSA-P patients tended to show higher tracer uptake in the lentiform nucleus. A multinomial logistic regression classified 33/34 patients into the correct clinical diagnosis group. Tracer uptake in the pons, cerebellar deep white matter, and striatum was closely associated with the presence of cerebellar and parkinsonian symptoms of MSA patients. The current study demonstrates that combined MAO-B and tau binding of THK5351 facilitates differential diagnosis of parkinsonian syndromes. Furthermore, our data indicate a correlation of MSA phenotype with [18F]-THK5351 uptake in certain brain regions, illustrating their relevance for the emergence of clinical symptoms and underlining the potential of THK5351 PET as a biomarker that correlates with pathological changes as well as with disease stage.
Collapse
Affiliation(s)
- Sonja Schönecker
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany
| | - Carla Palleis
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Technical University of Munich, Munich, Germany
| | - Elisabeth Schuh
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Julia Sauerbeck
- Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany
| | - Guido Rohrer
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Sonnenfeld
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katsutoshi Furukawa
- Division of Community Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Aiko Ishiki
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany
| | - Marianne Dieterich
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Kai Bötzel
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, University Hospital LMU Munich, Munich, Germany.,Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
13
|
Kreimerman I, Reyes AL, Paolino A, Pardo T, Porcal W, Ibarra M, Oliver P, Savio E, Engler H. Biological Assessment of a 18F-Labeled Sulforhodamine 101 in a Mouse Model of Alzheimer's Disease as a Potential Astrocytosis Marker. Front Neurosci 2019; 13:734. [PMID: 31379487 PMCID: PMC6646682 DOI: 10.3389/fnins.2019.00734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative diseases have mainly been associated with neuronal death. Recent investigations have shown that astroglia may modulate neuroinflammation in the early and late stages of the disease. [11C]Deuterodeprenyl ([11C]DED) is a tracer that has been used for reactive astrocyte detection in Alzheimer’s disease, Creutzfeldt–Jakob disease and amyotrophic lateral sclerosis, among others, with some limitations. To develop a new radiotracer for detecting astrocytosis and overcoming associated difficulties, we recently reported the synthesis of a sulfonamide derivative of Sulforhodamine 101 (SR101), labeled with 18F, namely SR101 N-(3-[18F]Fluoropropyl) sulfonamide ([18F]2B-SRF101). The red fluorescent dye SR101 has been used as a specific marker of astroglia in the neocortex of rodents using in vivo models. In the present work we performed a biological characterisation of the new tracer including biodistribution and micro-PET/computed tomography (CT) images. PET/CT studies with [11C]DED were also done to compare with [18F]2B-SRF101 in order to assess its potential as an astrocyte marker. Biodistribution studies with [18F]2B-SRF101 were carried out in C57BL6J black and transgenic (3xTg) mice. A hepatointestinal metabolization as well as the pharmacokinetic profile were determined, showing appropriate characteristics to become a PET diagnostic agent. Dynamic PET/CT studies were carried out with [18F]2B-SRF101 and [11C]DED to evaluate the distribution of both tracers in the brain. A significant difference in [18F]2B-SRF101 uptake was especially observed in the cortex and hippocampus, and it was higher in 3xTg mice than it was in the control group. These results suggested that [18F]2B-SRF101 is a promising candidate for more extensive evaluation as an astrocyte tracer. The difference observed for [18F]2B-SRF101 was not found in the case of [11C]DED. The comparative studies between [18F]2B-SRF101 and [11C]DED suggest that both tracers have different roles as astrocytosis markers in this animal model, and could provide different and complementary information at the same time. In this way, by means of a multitracer approach, useful information could be obtained for the staging of the disease.
Collapse
Affiliation(s)
- Ingrid Kreimerman
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Ana Laura Reyes
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Andrea Paolino
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Tania Pardo
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Williams Porcal
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay.,Department of Organic Chemistry, Faculty of Chemistry, University of the Republic (UdelaR), Montevideo, Uruguay
| | - Manuel Ibarra
- Pharmaceutical Sciences Department, Faculty of Chemistry, University of the Republic (UdelaR), Montevideo, Uruguay
| | - Patricia Oliver
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Eduardo Savio
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Henry Engler
- Radiopharmacy Department, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| |
Collapse
|
14
|
Guest FL. Early Detection and Treatment of Patients with Alzheimer's Disease: Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:295-317. [PMID: 30747429 DOI: 10.1007/978-3-030-05542-4_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease affects approximately 6% of people over the age of 65 years. It is characterized as chronic degeneration of cortical neurons, with loss of memory, cognition and executive functions. As the disease progresses, it is accompanied by accumulation of amyloid plaques and neurofibrillary tangles in key areas of the brain, leading to a loss of neurogenesis and synaptic plasticity in the hippocampus, along with changes in the levels of essential neurotransmitters such as acetylcholine and glutamate. Individuals with concomitant diseases such as depression, diabetes and cardiovascular disorders have a higher risk of developing Alzheimer's disease, and those who have a healthier diet and partake in regular exercise and intellectual stimulation have a lower risk of developing the disorder. This chapter describes the advances made in early diagnosis of Alzheimer's disease as this could help to improve outcomes for the patients by facilitating earlier treatment.
Collapse
Affiliation(s)
- Francesca L Guest
- Taunton and Somerset NHS Trust, Musgrove Park Hospital, Taunton, Somerset, UK.
| |
Collapse
|