101
|
Magaki SD, Vinters HV, Williams CK, Mareninov S, Khanlou N, Said J, Nemanim N, Gonzalez J, Morales JG, Singer EJ, Yong WH. Neuropathologic Findings in Elderly HIV-Positive Individuals. J Neuropathol Exp Neurol 2022; 81:565-576. [PMID: 35656871 DOI: 10.1093/jnen/nlac040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The elderly HIV-positive population is growing due to the widespread use of combination antiretroviral therapy (cART), but the effects of longstanding HIV infection on brain aging are unknown. A significant proportion of HIV-positive individuals develop HIV-associated neurocognitive disorder (HAND) even on cART, but the pathogenesis of HAND is unknown. Although neuroinflammation is postulated to play an important role in aging and neurodegenerative diseases such as Alzheimer disease (AD), it is unclear whether HIV accelerates aging or increases the risk for AD. We examined the brains of 9 elderly HIV-positive subjects on cART without co-infection by hepatitis C virus compared to 7 elderly HIV-negative subjects. Microglial and astrocyte activation and AD pathologic change in association with systemic comorbidities and neurocognitive assessment were evaluated. There was no difference in microglial or astrocyte activation between our HIV-positive and HIV-negative cohorts. One HIV-positive subject and 2 HIV-negative subjects demonstrated significant amyloid deposition, predominantly in the form of diffuse senile plaques, but these individuals were cognitively normal. Neurofibrillary tangles were sparse in the HIV-positive cohort. There was a high prevalence of cardiovascular comorbidities in all subjects. These findings suggest that multiple factors likely contribute to aging and cognitive impairment in elderly HIV-positive individuals on cART.
Collapse
Affiliation(s)
- Shino D Magaki
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Harry V Vinters
- Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Christopher K Williams
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sergey Mareninov
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Negar Khanlou
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jonathan Said
- Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Natasha Nemanim
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jessica Gonzalez
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jose G Morales
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - Elyse J Singer
- Department of Neurology, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| | - William H Yong
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, Ronald Reagan UCLA Medical Center and David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
102
|
Rajesh Y, Kanneganti TD. Innate Immune Cell Death in Neuroinflammation and Alzheimer's Disease. Cells 2022; 11:1885. [PMID: 35741014 PMCID: PMC9221514 DOI: 10.3390/cells11121885] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder molecularly characterized by the formation of amyloid β (Aβ) plaques and type 2 microtubule-associated protein (Tau) abnormalities. Multiple studies have shown that many of the brain's immunological cells, specifically microglia and astrocytes, are involved in AD pathogenesis. Cells of the innate immune system play an essential role in eliminating pathogens but also regulate brain homeostasis and AD. When activated, innate immune cells can cause programmed cell death through multiple pathways, including pyroptosis, apoptosis, necroptosis, and PANoptosis. The cell death often results in the release of proinflammatory cytokines that propagate the innate immune response and can eliminate Aβ plaques and aggregated Tau proteins. However, chronic neuroinflammation, which can result from cell death, has been linked to neurodegenerative diseases and can worsen AD. Therefore, the innate immune response must be tightly balanced to appropriately clear these AD-related structural abnormalities without inducing chronic neuroinflammation. In this review, we discuss neuroinflammation, innate immune responses, inflammatory cell death pathways, and cytokine secretion as they relate to AD. Therapeutic strategies targeting these innate immune cell death mechanisms will be critical to consider for future preventive or palliative treatments for AD.
Collapse
|
103
|
Microglia in Alzheimer’s Disease: A Favorable Cellular Target to Ameliorate Alzheimer’s Pathogenesis. Mediators Inflamm 2022; 2022:6052932. [PMID: 35693110 PMCID: PMC9184163 DOI: 10.1155/2022/6052932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Microglial cells serve as molecular sensors of the brain that play a role in physiological and pathological conditions. Under normal physiology, microglia are primarily responsible for regulating central nervous system homeostasis through the phagocytic clearance of redundant protein aggregates, apoptotic cells, damaged neurons, and synapses. Furthermore, microglial cells can promote and mitigate amyloid β phagocytosis and tau phosphorylation. Dysregulation of the microglial programming alters cellular morphology, molecular signaling, and secretory inflammatory molecules that contribute to various neurodegenerative disorders especially Alzheimer’s disease (AD). Furthermore, microglia are considered primary sources of inflammatory molecules and can induce or regulate a broad spectrum of cellular responses. Interestingly, in AD, microglia play a double-edged role in disease progression; for instance, the detrimental microglial effects increase in AD while microglial beneficiary mechanisms are jeopardized. Depending on the disease stages, microglial cells are expressed differently, which may open new avenues for AD therapy. However, the disease-related role of microglial cells and their receptors in the AD brain remain unclear. Therefore, this review represents the role of microglial cells and their involvement in AD pathogenesis.
Collapse
|
104
|
Gómez-Isla T, Frosch MP. Lesions without symptoms: understanding resilience to Alzheimer disease neuropathological changes. Nat Rev Neurol 2022; 18:323-332. [PMID: 35332316 PMCID: PMC10607925 DOI: 10.1038/s41582-022-00642-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
Abstract
Since the original description of amyloid-β plaques and tau tangles more than 100 years ago, these lesions have been considered the neuropathological hallmarks of Alzheimer disease (AD). The prevalence of plaques, tangles and dementia increases with age, and the lesions are considered to be causally related to the cognitive symptoms of AD. Current schemes for assessing AD lesion burden examine the distribution, abundance and characteristics of plaques and tangles at post mortem, yielding an estimate of the likelihood of cognitive impairment. Although this approach is highly predictive for most individuals, in some instances, a striking mismatch between lesions and symptoms can be observed. A small subset of individuals harbour a high burden of plaques and tangles at autopsy, which would be expected to have had devastating clinical consequences, but remain at their cognitive baseline, indicating 'resilience'. The study of these brains might provide the key to understanding the 'black box' between the accumulation of plaques and tangles and cognitive impairment, and show the way towards disease-modifying treatments for AD. In this Review, we begin by considering the heterogeneity of clinical manifestations associated with the presence of plaques and tangles, and then focus on insights derived from the rare yet informative individuals who display high amounts of amyloid and tau deposition in their brains (observed directly at autopsy) without manifesting dementia during life. The resilient response of these individuals to the gradual accumulation of plaques and tangles has potential implications for assessing an individual's risk of AD and for the development of interventions aimed at preserving cognition.
Collapse
Affiliation(s)
- Teresa Gómez-Isla
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA.
| | - Matthew P Frosch
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
105
|
Li X, Yan B, Du J, Xu S, Liu L, Pan C, Kang X, Zhu S. Recent Advances in Progresses and Prospects of IL-37 in Central Nervous System Diseases. Brain Sci 2022; 12:brainsci12060723. [PMID: 35741608 PMCID: PMC9221119 DOI: 10.3390/brainsci12060723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Interleukin-37 (IL-37) is an effective anti-inflammatory factor and acts through intracellular and extracellular pathways, inhibiting the effects of other inflammatory cytokines, such as IL-1β, IL-6, and tumor necrosis factor-α (TNF-α), thereby exerting powerful anti-inflammatory effects. In numerous recent studies, the anti-inflammatory effects of IL-37 have been described in many autoimmune diseases, colitis, and tumors. However, the current research on IL-37 in the field of the central nervous system (CNS) is not only less, but mainly for clinical research and little discussion of the mechanism. In this review, the role of IL-37 and its associated inflammatory factors in common CNS diseases are summarized, and their therapeutic potential in CNS diseases identified.
Collapse
Affiliation(s)
- Xinrui Li
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
| | - Bing Yan
- Department of Anesthesiology, Haining People’s Hospital, Haining 314499, China;
| | - Jin Du
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
- China Coast Guard Hospital of the People‘s Armed Police Force, Jiaxing 314000, China
| | - Shanshan Xu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
| | - Lu Liu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
| | - Caifei Pan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
- Correspondence: (X.K.); (S.Z.)
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.L.); (J.D.); (S.X.); (L.L.); (C.P.)
- Correspondence: (X.K.); (S.Z.)
| |
Collapse
|
106
|
Taddei RN, Sanchez-Mico MV, Bonnar O, Connors T, Gaona A, Denbow D, Frosch MP, Gómez-Isla T. Changes in glial cell phenotypes precede overt neurofibrillary tangle formation, correlate with markers of cortical cell damage, and predict cognitive status of individuals at Braak III-IV stages. Acta Neuropathol Commun 2022; 10:72. [PMID: 35534858 PMCID: PMC9082857 DOI: 10.1186/s40478-022-01370-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/10/2022] Open
Abstract
Clinico-pathological correlation studies show that some otherwise healthy elderly individuals who never developed cognitive impairment harbor a burden of Alzheimer's disease lesions (plaques and tangles) that would be expected to result in dementia. In the absence of comorbidities explaining such discrepancies, there is a need to identify other brain changes that meaningfully contribute to the cognitive status of an individual in the face of such burdens of plaques and tangles. Glial inflammatory responses, a universal phenomenon in symptomatic AD, show robust association with degree of cognitive impairment, but their significance in early tau pathology stages and contribution to the trajectory of cognitive decline at an individual level remain widely unexplored. We studied 55 brains from individuals at intermediate stages of tau tangle pathology (Braak III-IV) with diverging antemortem cognition (demented vs. non-demented, here termed `resilient'), and age-matched cognitively normal controls (Braak 0-II). We conducted quantitative assessments of amyloid and tau lesions, cellular vulnerability markers, and glial phenotypes in temporal pole (Braak III-IV region) and visual cortex (Braak V-VI region) using artificial-intelligence based semiautomated quantifications. We found distinct glial responses with increased proinflammatory and decreased homeostatic markers, both in regions with tau tangles (temporal pole) and without overt tau deposits (visual cortex) in demented but not in resilient. These changes were significantly associated with markers of cortical cell damage. Similar phenotypic glial changes were detected in the white matter of demented but not resilient and were associated with higher burden of overlying cortical cellular damage in regions with and without tangles. Our data suggest that changes in glial phenotypes in cortical and subcortical regions represent an early phenomenon that precedes overt tau deposition and likely contributes to cell damage and loss of brain function predicting the cognitive status of individuals at intermediate stages of tau aggregate burden (Braak III-IV).
Collapse
Affiliation(s)
- Raquel N Taddei
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- Department of Neurology, Dementia Research Institute, University College London, London, UK
| | - Maria V Sanchez-Mico
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Orla Bonnar
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Theresa Connors
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Angelica Gaona
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Dominique Denbow
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Matthew P Frosch
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA.
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA.
| |
Collapse
|
107
|
Yuste-Checa P, Bracher A, Hartl FU. The chaperone Clusterin in neurodegeneration-friend or foe? Bioessays 2022; 44:e2100287. [PMID: 35521968 DOI: 10.1002/bies.202100287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022]
Abstract
Fibrillar protein aggregates are the pathological hallmark of a group of age-dependent neurodegenerative conditions, including Alzheimer's and Parkinson's disease. Aggregates of the microtubule-associated protein Tau are observed in Alzheimer's disease and primary tauopathies. Tau pathology propagates from cell to cell in a prion-like process that is likely subject to modulation by extracellular chaperones such as Clusterin. We recently reported that Clusterin delayed Tau fibril formation but enhanced the activity of Tau oligomers to seed aggregation of endogenous Tau in a cellular model. In contrast, Clusterin inhibited the propagation of α-Synuclein aggregates associated with Parkinson's disease. These findings raise the possibility of a mechanistic link between Clusterin upregulation observed in Alzheimer's disease and the progression of Tau pathology. Here we review the diverse functions of Clusterin in the pathogenesis of neurodegenerative diseases, focusing on evidence that Clusterin may act either as a suppressor or enhancer of pathology.
Collapse
Affiliation(s)
- Patricia Yuste-Checa
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Andreas Bracher
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
108
|
Lazic A, Balint V, Stanisavljevic Ninkovic D, Peric M, Stevanovic M. Reactive and Senescent Astroglial Phenotypes as Hallmarks of Brain Pathologies. Int J Mol Sci 2022; 23:ijms23094995. [PMID: 35563385 PMCID: PMC9100382 DOI: 10.3390/ijms23094995] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes, as the most abundant glial cells in the central nervous system, are tightly integrated into neural networks and participate in numerous aspects of brain physiology and pathology. They are the main homeostatic cells in the central nervous system, and the loss of astrocyte physiological functions and/or gain of pro-inflammatory functions, due to their reactivation or cellular senescence, can have profound impacts on the surrounding microenvironment with pathological outcomes. Although the importance of astrocytes is generally recognized, and both senescence and reactive astrogliosis have been extensively reviewed independently, there are only a few comparative overviews of these complex processes. In this review, we summarize the latest data regarding astrocyte reactivation and senescence, and outline similarities and differences between these phenotypes from morphological, functional, and molecular points of view. A special focus has been given to neurodegenerative diseases, where these phenotypic alternations of astrocytes are significantly implicated. We also summarize current perspectives regarding new advances in model systems based on astrocytes as well as data pointing to these glial cells as potential therapeutic targets.
Collapse
Affiliation(s)
- Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Correspondence:
| | - Vanda Balint
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Mina Peric
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11001 Belgrade, Serbia
| |
Collapse
|
109
|
de Oliveira P, Cella C, Locker N, Ravindran KKG, Mendis A, Wafford K, Gilmour G, Dijk DJ, Winsky-Sommerer R. Improved Sleep, Memory, and Cellular Pathological Features of Tauopathy, Including the NLRP3 Inflammasome, after Chronic Administration of Trazodone in rTg4510 Mice. J Neurosci 2022; 42:3494-3509. [PMID: 35273086 PMCID: PMC9034788 DOI: 10.1523/jneurosci.2162-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 12/02/2022] Open
Abstract
Several cellular pathways contribute to neurodegenerative tauopathy-related disorders. Microglial activation, a major component of neuroinflammation, is an early pathologic hallmark that correlates with cognitive decline, while the unfolded protein response (UPR) contributes to synaptic pathology. Sleep disturbances are prevalent in tauopathies and may also contribute to disease progression. Few studies have investigated whether manipulations of sleep influence cellular pathologic and behavioral features of tauopathy. We investigated whether trazodone, a licensed antidepressant with hypnotic efficacy in dementia, can reduce disease-related cellular pathways and improve memory and sleep in male rTg4510 mice with a tauopathy-like phenotype. In a 9 week dosing regimen, trazodone decreased microglial NLRP3 inflammasome expression and phosphorylated p38 mitogen-activated protein kinase levels, which correlated with the NLRP3 inflammasome, the UPR effector ATF4, and total tau levels. Trazodone reduced theta oscillations during rapid eye movement (REM) sleep and enhanced REM sleep duration. Olfactory memory transiently improved, and memory performance correlated with REM sleep duration and theta oscillations. These findings on the effects of trazodone on the NLRP3 inflammasome, the unfolded protein response and behavioral hallmarks of dementia warrant further studies on the therapeutic value of sleep-modulating compounds for tauopathies.SIGNIFICANCE STATEMENT Dementia and associated behavioral symptoms such as memory loss and sleep disturbance are debilitating. Identifying treatments that alleviate symptoms and concurrently target cellular pathways contributing to disease progression is paramount for the patients and their caregivers. Here we show that a chronic treatment with trazodone, an antidepressant with positive effects on sleep, has beneficial effects on several cellular pathways contributing to neuroinflammation and tau pathology, in tauopathy-like rTg4510 mice. Trazodone also improved rapid eye movement (REM) sleep, the slowing of brain oscillations, and olfactory memory disturbances, which are all early symptoms observed in Alzheimer's disease. Thus, trazodone and compounds with REM sleep-promoting properties may represent a promising treatment approach to reduce the early symptoms of tauopathy and slow down disease progression.
Collapse
Affiliation(s)
- Paula de Oliveira
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Claire Cella
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Nicolas Locker
- Department of Microbial and Cellular Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7TE, United Kingdom
| | - Kiran K G Ravindran
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London, London W12 0BZ and University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Agampodi Mendis
- Surrey Clinical Trials Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Keith Wafford
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Gary Gilmour
- Lilly Research Centre, Eli Lilly and Company, Windlesham GU20 6PH, United Kingdom
| | - Derk-Jan Dijk
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London, London W12 0BZ and University of Surrey, Guildford GU2 7XP, United Kingdom
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XP, United Kingdom
| |
Collapse
|
110
|
Alzheimer’s Disease and Tau Self-Assembly: In the Search of the Missing Link. Int J Mol Sci 2022; 23:ijms23084192. [PMID: 35457009 PMCID: PMC9032712 DOI: 10.3390/ijms23084192] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative disease characterized by progressive cognitive impairment, apathy, and neuropsychiatric disorders. Two main pathological hallmarks have been described: neurofibrillary tangles, consisting of tau oligomers (hyperphosphorylated tau) and Aβ plaques. The influence of protein kinases and phosphatases on the hyperphosphorylation of tau is already known. Hyperphosphorylated tau undergoes conformational changes that promote its self-assembly. However, the process involving these mechanisms is yet to be elucidated. In vitro recombinant tau can be aggregated by the action of polyanions, such as heparin, arachidonic acid, and more recently, the action of polyphosphates. However, how that process occurs in vivo is yet to be understood. In this review, searching the most accurate and updated literature on the matter, we focus on the precise molecular events linking tau modifications, its misfolding and the initiation of its pathological self-assembly. Among these, we can identify challenges regarding tau phosphorylation, the link between tau heteroarylations and the onset of its self-assembly, as well as the possible metabolic pathways involving natural polyphosphates, that may play a role in tau self-assembly.
Collapse
|
111
|
Gourmaud S, Stewart DA, Irwin DJ, Roberts N, Barbour AJ, Eberwine G, O’Brien WT, Vassar R, Talos DM, Jensen FE. The role of mTORC1 activation in seizure-induced exacerbation of Alzheimer's disease. Brain 2022; 145:324-339. [PMID: 34264340 PMCID: PMC9126019 DOI: 10.1093/brain/awab268] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/04/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
The risk of seizures is 10-fold higher in patients with Alzheimer's disease than the general population, yet the mechanisms underlying this susceptibility and the effects of these seizures are poorly understood. To elucidate the proposed bidirectional relationship between Alzheimer's disease and seizures, we studied human brain samples (n = 34) from patients with Alzheimer's disease and found that those with a history of seizures (n = 14) had increased amyloid-β and tau pathology, with upregulation of the mechanistic target of rapamycin (mTOR) pathway, compared with patients without a known history of seizures (n = 20). To establish whether seizures accelerate the progression of Alzheimer's disease, we induced chronic hyperexcitability in the five times familial Alzheimer's disease mouse model by kindling with the chemoconvulsant pentylenetetrazol and observed that the mouse model exhibited more severe seizures than the wild-type. Furthermore, kindled seizures exacerbated later cognitive impairment, Alzheimer's disease neuropathology and mTOR complex 1 activation. Finally, we demonstrated that the administration of the mTOR inhibitor rapamycin following kindled seizures rescued enhanced remote and long-term memory deficits associated with earlier kindling and prevented seizure-induced increases in Alzheimer's disease neuropathology. These data demonstrated an important link between chronic hyperexcitability and progressive Alzheimer's disease pathology and suggest a mechanism whereby rapamycin may serve as an adjunct therapy to attenuate progression of the disease.
Collapse
Affiliation(s)
- Sarah Gourmaud
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David A Stewart
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Duke University School of Medicine, Durham, NC 27708, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Frontotemporal Degeneration Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas Roberts
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron J Barbour
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace Eberwine
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T O’Brien
- Neurobehavior Testing Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Delia M Talos
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
112
|
Zhou X, Venigalla M, Raju R, Münch G. Pharmacological considerations for treating neuroinflammation with curcumin in Alzheimer's disease. J Neural Transm (Vienna) 2022; 129:755-771. [PMID: 35294663 DOI: 10.1007/s00702-022-02480-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/19/2022] [Indexed: 12/14/2022]
Abstract
Prof. Dr. Peter Riederer, the former Head of the Neurochemistry Department of the Psychiatry and Psychotherapy Clinic at the University of Würzburg (Germany), has been one of the pioneers of research into oxidative stress in Parkinson's and Alzheimer's disease (AD). This review will outline how his scientific contribution to the field has opened a new direction for AD treatment beyond "plaques and tangles". In the 1990s, Prof. Riederer was one of the first scientists who proposed oxidative stress and neuroinflammation as one of the major contributors to Alzheimer's disease, despite the overwhelming support for the "amyloid-only" hypothesis at the time, which postulated that the sole and only cause of AD is β-amyloid. His group also highlighted the role of advanced glycation end products, sugar and dicarbonyl-derived protein modifications, which crosslink proteins into insoluble aggregates and potent pro-inflammatory activators of microglia. For the treatment of chronic neuroinflammation, he and his group suggested that the most appropriate drug class would be cytokine-suppressive anti-inflammatory drugs (CSAIDs) which have a broader anti-inflammatory action range than conventional non-steroidal anti-inflammatory drugs. One of the most potent CSAIDs is curcumin, but it suffers from a variety of pharmacokinetic disadvantages including low bioavailability, which might have tainted many human clinical trials. Although a variety of oral formulations with increased bioavailability have been developed, curcumin's absorption after oral delivery is too low to reach therapeutic concentrations in the micromolar range in the systemic circulation and the brain. This review will conclude with evidence that rectally applied suppositories might be the best alternatives to oral medications, as this route will be able to evade first-pass metabolism in the liver and achieve high concentrations of curcumin in plasma and tissues, including the brain.
Collapse
Affiliation(s)
- Xian Zhou
- NICM Health Research Institute, Western Sydney University, 158-160 Hawkesbury Rd, Westmead, NSW, 2145, Australia
| | - Madhuri Venigalla
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Ritesh Raju
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia.
| |
Collapse
|
113
|
Thuraisingham RA. A kinetic scheme to examine the role of glial cells in the pathogenesis of Alzheimer's disease. Metab Brain Dis 2022; 37:801-805. [PMID: 35032278 DOI: 10.1007/s11011-022-00902-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/03/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that leads to severe impairments in cognitive functions including memory and learning. An improved kinetic model is proposed here to understand the pathogenesis of AD in particular the role of glial cells in the presence of amyloid plaques and neurofibrillary tangles (NFTs). The kinetic model describes the production of activated microglia and astroglia. It involves two rate equations and incorporates the dual role of these glial cells which can function as neuroprotective and as neurotoxic cells. Examination of the steady state solutions of the model predicts an increase in population of these glial cells as (AD) progresses, and that this continues to increase linearly even after the amyloid population has reached a plateau.This is in agreement with experimental data. Limiting AD to the effect of amyloid peptides alone is incorrect and the role of neurofibrillary tangles, clearance rate of dead neurons and neuroinflammation from glial cells are vital and must be included in understanding the pathogenesis of AD. The study shows that increasing the clearance of dead neurons and use of any method to deactivate the glial cells will diminish the progression of AD.
Collapse
|
114
|
Garland EF, Hartnell IJ, Boche D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front Neurosci 2022; 16:824888. [PMID: 35250459 PMCID: PMC8888691 DOI: 10.3389/fnins.2022.824888] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia and astrocytes play essential roles in the central nervous system contributing to many functions including homeostasis, immune response, blood-brain barrier maintenance and synaptic support. Evidence has emerged from experimental models of glial communication that microglia and astrocytes influence and coordinate each other and their effects on the brain environment. However, due to the difference in glial cells between humans and rodents, it is essential to confirm the relevance of these findings in human brains. Here, we aim to review the current knowledge on microglia-astrocyte crosstalk in humans, exploring novel methodological techniques used in health and disease conditions. This will include an in-depth look at cell culture and iPSCs, post-mortem studies, imaging and fluid biomarkers, genetics and transcriptomic data. In this review, we will discuss the advantages and limitations of these methods, highlighting the understanding these methods have brought the field on these cells communicative abilities, and the knowledge gaps that remain.
Collapse
Affiliation(s)
| | | | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
115
|
Muñoz-Castro C, Noori A, Magdamo CG, Li Z, Marks JD, Frosch MP, Das S, Hyman BT, Serrano-Pozo A. Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer's disease. J Neuroinflammation 2022; 19:30. [PMID: 35109872 PMCID: PMC8808995 DOI: 10.1186/s12974-022-02383-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Astrocytes and microglia react to Aβ plaques, neurofibrillary tangles, and neurodegeneration in the Alzheimer's disease (AD) brain. Single-nuclei and single-cell RNA-seq have revealed multiple states or subpopulations of these glial cells but lack spatial information. We have developed a methodology of cyclic multiplex fluorescent immunohistochemistry on human postmortem brains and image analysis that enables a comprehensive morphological quantitative characterization of astrocytes and microglia in the context of their spatial relationships with plaques and tangles. METHODS Single FFPE sections from the temporal association cortex of control and AD subjects were subjected to 8 cycles of multiplex fluorescent immunohistochemistry, including 7 astroglial, 6 microglial, 1 neuronal, Aβ, and phospho-tau markers. Our analysis pipeline consisted of: (1) image alignment across cycles; (2) background subtraction; (3) manual annotation of 5172 ALDH1L1+ astrocytic and 6226 IBA1+ microglial profiles; (4) local thresholding and segmentation of profiles; (5) machine learning on marker intensity data; and (6) deep learning on image features. RESULTS Spectral clustering identified three phenotypes of astrocytes and microglia, which we termed "homeostatic," "intermediate," and "reactive." Reactive and, to a lesser extent, intermediate astrocytes and microglia were closely associated with AD pathology (≤ 50 µm). Compared to homeostatic, reactive astrocytes contained substantially higher GFAP and YKL-40, modestly elevated vimentin and TSPO as well as EAAT1, and reduced GS. Intermediate astrocytes had markedly increased EAAT2, moderately increased GS, and intermediate GFAP and YKL-40 levels. Relative to homeostatic, reactive microglia showed increased expression of all markers (CD68, ferritin, MHC2, TMEM119, TSPO), whereas intermediate microglia exhibited increased ferritin and TMEM119 as well as intermediate CD68 levels. Machine learning models applied on either high-plex signal intensity data (gradient boosting machines) or directly on image features (convolutional neural networks) accurately discriminated control vs. AD diagnoses at the single-cell level. CONCLUSIONS Cyclic multiplex fluorescent immunohistochemistry combined with machine learning models holds promise to advance our understanding of the complexity and heterogeneity of glial responses as well as inform transcriptomics studies. Three distinct phenotypes emerged with our combination of markers, thus expanding the classic binary "homeostatic vs. reactive" classification to a third state, which could represent "transitional" or "resilient" glia.
Collapse
Affiliation(s)
- Clara Muñoz-Castro
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Sevilla, Spain
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Ayush Noori
- Harvard College, Boston, MA, 02138, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Colin G Magdamo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Zhaozhi Li
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Jordan D Marks
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, 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 Street, Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
116
|
Silburt J, Aubert I. MORPHIOUS: an unsupervised machine learning workflow to detect the activation of microglia and astrocytes. J Neuroinflammation 2022; 19:24. [PMID: 35093113 PMCID: PMC8800241 DOI: 10.1186/s12974-021-02376-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background In conditions of brain injury and degeneration, defining microglial and astrocytic activation using cellular markers alone remains a challenging task. We developed the MORPHIOUS software package, an unsupervised machine learning workflow which can learn the morphologies of non-activated astrocytes and microglia, and from this information, infer clusters of microglial and astrocytic activation in brain tissue. Methods MORPHIOUS combines a one-class support vector machine with the density-based spatial clustering of applications with noise (DBSCAN) algorithm to identify clusters of microglial and astrocytic activation. Here, activation was triggered by permeabilizing the blood–brain barrier (BBB) in the mouse hippocampus using focused ultrasound (FUS). At 7 day post-treatment, MORPHIOUS was applied to evaluate microglial and astrocytic activation in histological tissue. MORPHIOUS was further evaluated on hippocampal sections of TgCRND8 mice, a model of amyloidosis that is prone to microglial and astrocytic activation. Results MORPHIOUS defined two classes of microglia, termed focal and proximal, that are spatially adjacent to the activating stimulus. Focal and proximal microglia demonstrated activity-associated features, including increased levels of ionized calcium-binding adapter molecule 1 expression, enlarged soma size, and deramification. MORPHIOUS further identified clusters of astrocytes characterized by activity-related changes in glial fibrillary acidic protein expression and branching. To validate these classifications following FUS, co-localization with activation markers were assessed. Focal and proximal microglia co-localized with the transforming growth factor beta 1, while proximal astrocytes co-localized with Nestin. In TgCRND8 mice, microglial and astrocytic activation clusters were found to correlate with amyloid-β plaque load. Thus, by only referencing control microglial and astrocytic morphologies, MORPHIOUS identified regions of interest corresponding to microglial and astrocytic activation. Conclusions Overall, our algorithm is a reliable and sensitive method for characterizing microglial and astrocytic activation following FUS-induced BBB permeability and in animal models of neurodegeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02376-9.
Collapse
|
117
|
Gouilly D, Saint-Aubert L, Ribeiro MJ, Salabert AS, Tauber C, Péran P, Arlicot N, Pariente J, Payoux P. Neuroinflammation PET imaging of the translocator protein (TSPO) in Alzheimer's disease: an update. Eur J Neurosci 2022; 55:1322-1343. [PMID: 35083791 DOI: 10.1111/ejn.15613] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is a significant contributor to Alzheimer's disease (AD). Until now, PET imaging of the translocator protein (TSPO) has been widely used to depict the neuroimmune endophenotype of AD. The aim of this review was to provide an update to the results from 2018 and to advance the characterization of the biological basis of TSPO imaging in AD by re-examining TSPO function and expression and the methodological aspects of interest. Although the biological basis of the TSPO PET signal is obviously related to microglia and astrocytes in AD, the observed process remains uncertain and might not be directly related to neuroinflammation. Further studies are required to re-examine the cellular significance underlying a variation in the PET signal in AD and how it can be impacted by a disease-modifying treatment.
Collapse
Affiliation(s)
- Dominique Gouilly
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Laure Saint-Aubert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Maria-Joao Ribeiro
- Department of Nuclear Medicine, CHU, Tours, France.,UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| | | | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Jérémie Pariente
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU, Toulouse, France.,Center of Clinical Investigations (CIC1436), CHU, Toulouse, France
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| |
Collapse
|
118
|
Hastings N, Kuan WL, Osborne A, Kotter MRN. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant 2022; 31:9636897221105499. [PMID: 35770772 PMCID: PMC9251977 DOI: 10.1177/09636897221105499] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson's, Alzheimer's, and Huntington's diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Collapse
Affiliation(s)
- Nataly Hastings
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R N Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
119
|
Patel H, Martinez P, Perkins A, Taylor X, Jury N, McKinzie D, Lasagna-Reeves CA. Pathological tau and reactive astrogliosis are associated with distinct functional deficits in a mouse model of tauopathy. Neurobiol Aging 2022; 109:52-63. [PMID: 34655981 PMCID: PMC8671336 DOI: 10.1016/j.neurobiolaging.2021.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/12/2021] [Accepted: 09/05/2021] [Indexed: 01/03/2023]
Abstract
Pathological aggregation of tau and neuroinflammatory changes mark the clinical course of Alzheimer's disease and related tauopathies. To understand the correlation between these pathological hallmarks and functional deficits, we assessed behavioral and physiological deficits in the PS19 mouse model, a broadly utilized model of tauopathy. At 9 months, PS19 mice have characteristic hyperactive behavior, a decline in motor strength, and deterioration in physiological conditions marked by lower body temperature, reduced body weight, and an increase in measures of frailty. Correlation of these deficits with different pathological hallmarks revealed that pathological tau species, characterized by soluble p-tau species, and tau seeding bioactivity correlated with impairment in grip strength and thermal regulation. On the other hand, astrocyte reactivity showed a positive correlation with the hyperactive behavior of the PS19 mice. These results suggest that a diverse spectrum of soluble pathological tau species could be responsible for different symptoms and that neuroinflammation could contribute to functional deficits independently from tau pathology. These observations enhance the necessity of a multi-targeted approach for the treatment of neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Henika Patel
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Pablo Martinez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Abigail Perkins
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xavier Taylor
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nur Jury
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - David McKinzie
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cristian A. Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA,Corresponding author: Cristian A. Lasagna-Reeves, Ph.D., Indiana University School of Medicine, The Stark Neurosciences Research Institute, Neurosciences Research Building 214G, 320 West 15th Street, Indianapolis, IN, 46202, Office: (317) 274-7830,
| |
Collapse
|
120
|
Belkozhayev AM, Al-Yozbaki M, George A, Niyazova RY, Sharipov KO, Byrne LJ, Wilson CM. Extracellular Vesicles, Stem Cells and the Role of miRNAs in Neurodegeneration. Curr Neuropharmacol 2022; 20:1450-1478. [PMID: 34414870 PMCID: PMC9881087 DOI: 10.2174/1570159x19666210817150141] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/16/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
There are different modalities of intercellular communication governed by cellular homeostasis. In this review, we will explore one of these forms of communication called extracellular vesicles (EVs). These vesicles are released by all cells in the body and are heterogeneous in nature. The primary function of EVs is to share information through their cargo consisting of proteins, lipids and nucleic acids (mRNA, miRNA, dsDNA etc.) with other cells, which have a direct consequence on their microenvironment. We will focus on the role of EVs of mesenchymal stem cells (MSCs) in the nervous system and how these participate in intercellular communication to maintain physiological function and provide neuroprotection. However, deregulation of this same communication system could play a role in several neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, multiple sclerosis, prion disease and Huntington's disease. The release of EVs from a cell provides crucial information to what is happening inside the cell and thus could be used in diagnostics and therapy. We will discuss and explore new avenues for the clinical applications of using engineered MSC-EVs and their potential therapeutic benefit in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Ayaz M. Belkozhayev
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Minnatallah Al-Yozbaki
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Alex George
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
- Jubilee Centre for Medical Research, Jubilee Mission Medical College & Research Institute, Thrissur, Kerala, India
| | - Raigul Ye Niyazova
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
| | - Kamalidin O. Sharipov
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Lee J. Byrne
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Cornelia M. Wilson
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| |
Collapse
|
121
|
Gonzalez-Rodriguez M, Villar-Conde S, Astillero-Lopez V, Villanueva-Anguita P, Ubeda-Banon I, Flores-Cuadrado A, Martinez-Marcos A, Saiz-Sanchez D. Neurodegeneration and Astrogliosis in the Human CA1 Hippocampal Subfield Are Related to hsp90ab1 and bag3 in Alzheimer's Disease. Int J Mol Sci 2021; 23:165. [PMID: 35008592 PMCID: PMC8745315 DOI: 10.3390/ijms23010165] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disorder, is characterized by executive dysfunction and memory impairment mediated by the accumulation of extracellular amyloid-β peptide (Aβ) and intracellular hyperphosphorylated tau protein. The hippocampus (HIPP) is essential for memory formation and is involved in early stages of disease. In fact, hippocampal atrophy is used as an early biomarker of neuronal injury and to evaluate disease progression. It is not yet well-understood whether changes in hippocampal volume are due to neuronal or glial loss. The aim of the study was to assess hippocampal atrophy and/or gliosis using unbiased stereological quantification and to obtain hippocampal proteomic profiles related to neurodegeneration and gliosis. Hippocampal volume measurement, stereological quantification of NeuN-, Iba-1- and GFAP-positive cells, and sequential window acquisition of all theoretical mass spectrometry (SWATH-MS) analysis were performed in AD and non-AD cases. Reduced hippocampal volume was identified using the Cavalieri probe, particularly in the CA1 region, where it correlated with neuronal loss and astrogliosis. A total of 102 downregulated and 47 upregulated proteins were identified in the SWATH-MS analysis after restrictive filtering based on an FC > 1.5 and p value < 0.01. The Hsp90 family of chaperones, particularly BAG3 and HSP90AB1, are closely related to astrocytes, indicating a possible role in degrading Aβ and tau through chaperone-mediated autophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alino Martinez-Marcos
- CRIB, Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (M.G.-R.); (S.V.-C.); (V.A.-L.); (P.V.-A.); (I.U.-B.); (A.F.-C.)
| | - Daniel Saiz-Sanchez
- CRIB, Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain; (M.G.-R.); (S.V.-C.); (V.A.-L.); (P.V.-A.); (I.U.-B.); (A.F.-C.)
| |
Collapse
|
122
|
Pereira JB, Janelidze S, Smith R, Mattsson-Carlgren N, Palmqvist S, Teunissen CE, Zetterberg H, Stomrud E, Ashton NJ, Blennow K, Hansson O. Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer's disease. Brain 2021; 144:3505-3516. [PMID: 34259835 PMCID: PMC8677538 DOI: 10.1093/brain/awab223] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/24/2021] [Accepted: 06/05/2021] [Indexed: 11/13/2022] Open
Abstract
Although recent clinical trials targeting amyloid-β in Alzheimer's disease have shown promising results, there is increasing evidence suggesting that understanding alternative disease pathways that interact with amyloid-β metabolism and amyloid pathology might be important to halt the clinical deterioration. In particular, there is evidence supporting a critical role of astroglial activation and astrocytosis in Alzheimer's disease. However, so far, no studies have assessed whether astrocytosis is independently related to either amyloid-β or tau pathology in vivo. To address this question, we determined the levels of the astrocytic marker GFAP in plasma and CSF of 217 amyloid-β-negative cognitively unimpaired individuals, 71 amyloid-β-positive cognitively unimpaired individuals, 78 amyloid-β-positive cognitively impaired individuals, 63 amyloid-β-negative cognitively impaired individuals and 75 patients with a non-Alzheimer's disease neurodegenerative disorder from the Swedish BioFINDER-2 study. Participants underwent longitudinal amyloid-β (18F-flutemetamol) and tau (18F-RO948) PET as well as cognitive testing. We found that plasma GFAP concentration was significantly increased in all amyloid-β-positive groups compared with participants without amyloid-β pathology (P < 0.01). In addition, there were significant associations between plasma GFAP with higher amyloid-β-PET signal in all amyloid-β-positive groups, but also in cognitively normal individuals with normal amyloid-β values (P < 0.001), which remained significant after controlling for tau-PET signal. Furthermore, plasma GFAP could predict amyloid-β-PET positivity with an area under the curve of 0.76, which was greater than the performance achieved by CSF GFAP (0.69) and other glial markers (CSF YKL-40: 0.64, soluble TREM2: 0.71). Although correlations were also observed between tau-PET and plasma GFAP, these were no longer significant after controlling for amyloid-β-PET. In contrast to plasma GFAP, CSF GFAP concentration was significantly increased in non-Alzheimer's disease patients compared to other groups (P < 0.05) and correlated with amyloid-β-PET only in amyloid-β-positive cognitively impaired individuals (P = 0.005). Finally, plasma GFAP was associated with both longitudinal amyloid-β-PET and cognitive decline, and mediated the effect of amyloid-β-PET on tau-PET burden, suggesting that astrocytosis secondary to amyloid-β aggregation might promote tau accumulation. Altogether, these findings indicate that plasma GFAP is an early marker associated with brain amyloid-β pathology but not tau aggregation, even in cognitively normal individuals with a normal amyloid-β status. This suggests that plasma GFAP should be incorporated in current hypothetical models of Alzheimer's disease pathogenesis and be used as a non-invasive and accessible tool to detect early astrocytosis secondary to amyloid-β pathology.
Collapse
Affiliation(s)
- Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Nicholas J Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
123
|
Role of IL-33/ST2 Axis in Chronic Inflammatory Neurological Disorderss. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2021. [DOI: 10.2478/sjecr-2020-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Interleukin-33 (IL-33) is a member of IL-1 family of cytokines, produced constitutively by fibroblasts, endothelial cells, and epithelial cells. IL-33 can be released passively from cells during tissue damage and cell necrosis, suggesting that it may act as an alarmin. Function of IL-33 is mediated by its interaction with ST2 molecule that is expressed on many immune cells: Th2 lymphocytes, NK, NKT and mast cells, monocytes, dendritic cells and granulocytes. IL-33/ST2 pathway plays, often dual, roles in different physiological and inflammatory processes, mediating both, pathological immune responses and tissue repair. Expression of IL-33 in the central nervous system (CNS) is significantly enhanced during various pathological processes, indicating its important role in the pathogenesis of neurological inflammatory and degenerative diseases. In this review the biological features, expression of IL-33 and its ligand ST2 in CNS, and the role of IL- 33/ST2 pathway in development of Alzheimer’s disease and multiple sclerosis are discussed.
Collapse
|
124
|
Staurenghi E, Giannelli S, Testa G, Sottero B, Leonarduzzi G, Gamba P. Cholesterol Dysmetabolism in Alzheimer's Disease: A Starring Role for Astrocytes? Antioxidants (Basel) 2021; 10:antiox10121890. [PMID: 34943002 PMCID: PMC8750262 DOI: 10.3390/antiox10121890] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/19/2023] Open
Abstract
In recent decades, the impairment of cholesterol metabolism in the pathogenesis of Alzheimer’s disease (AD) has been intensively investigated, and it has been recognized to affect amyloid β (Aβ) production and clearance, tau phosphorylation, neuroinflammation and degeneration. In particular, the key role of cholesterol oxidation products, named oxysterols, has emerged. Brain cholesterol metabolism is independent from that of peripheral tissues and it must be preserved in order to guarantee cerebral functions. Among the cells that help maintain brain cholesterol homeostasis, astrocytes play a starring role since they deliver de novo synthesized cholesterol to neurons. In addition, other physiological roles of astrocytes are to modulate synaptic transmission and plasticity and support neurons providing energy. In the AD brain, astrocytes undergo significant morphological and functional changes that contribute to AD onset and development. However, the extent of this contribution and the role played by oxysterols are still unclear. Here we review the current understanding of the physiological role exerted by astrocytes in the brain and their contribution to AD pathogenesis. In particular, we focus on the impact of cholesterol dysmetabolism on astrocyte functions suggesting new potential approaches to develop therapeutic strategies aimed at counteracting AD development.
Collapse
|
125
|
Garcia-Romeu A, Darcy S, Jackson H, White T, Rosenberg P. Psychedelics as Novel Therapeutics in Alzheimer's Disease: Rationale and Potential Mechanisms. Curr Top Behav Neurosci 2021; 56:287-317. [PMID: 34734390 DOI: 10.1007/7854_2021_267] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Serotonin 2A receptor (5-HT2AR) agonist "classic psychedelics" are drawing increasing interest as potential mental health treatments. Recent work suggests psychedelics can exert persisting anxiolytic and antidepressant effects lasting up to several months after a single administration. Data indicate acute subjective drug effects as important psychological factors involved in observed therapeutic benefits. Additionally, animal models have shown an important role for 5-HT2AR agonists in modulating learning and memory function with relevance for Alzheimer's Disease (AD) and related dementias. A number of biological mechanisms of action are under investigation to elucidate 5-HT2AR agonists' therapeutic potential, including enhanced neuroplasticity, anti-inflammatory effects, and alterations in brain functional connectivity. These diverse lines of research are reviewed here along with a discussion of AD pathophysiology and neuropsychiatric symptoms to highlight classic psychedelics as potential novel pharmacotherapies for patients with AD. Human clinical research suggests a possible role for high-dose psychedelic administration in symptomatic treatment of depressed mood and anxiety in early-stage AD. Preclinical data indicate a potential for low- or high-dose psychedelic treatment regimens to slow or reverse brain atrophy, enhance cognitive function, and slow progression of AD. In conclusion, rationale and potential approaches for preliminary research with psychedelics in patients with AD are presented, and ramifications of this line of investigation for development of novel AD treatments are discussed.
Collapse
Affiliation(s)
- Albert Garcia-Romeu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sean Darcy
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hillary Jackson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Toni White
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Memory and Alzheimer's Treatment Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Memory and Alzheimer's Treatment Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
126
|
Ulland TK, Ewald AC, Knutson AO, Marino KM, Smith SMC, Watters JJ. Alzheimer's Disease, Sleep Disordered Breathing, and Microglia: Puzzling out a Common Link. Cells 2021; 10:2907. [PMID: 34831129 PMCID: PMC8616348 DOI: 10.3390/cells10112907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/14/2022] Open
Abstract
Sleep Disordered Breathing (SDB) and Alzheimer's Disease (AD) are strongly associated clinically, but it is unknown if they are mechanistically associated. Here, we review data covering both the cellular and molecular responses in SDB and AD with an emphasis on the overlapping neuroimmune responses in both diseases. We extensively discuss the use of animal models of both diseases and their relative utilities in modeling human disease. Data presented here from mice exposed to intermittent hypoxia indicate that microglia become more activated following exposure to hypoxia. This also supports the idea that intermittent hypoxia can activate the neuroimmune system in a manner like that seen in AD. Finally, we highlight similarities in the cellular and neuroimmune responses between SDB and AD and propose that these similarities may lead to a pathological synergy between SDB and AD.
Collapse
Affiliation(s)
- Tyler K. Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Andrew O. Knutson
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Kaitlyn M. Marino
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI 53705, USA; (T.K.U.); (K.M.M.)
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Stephanie M. C. Smith
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| | - Jyoti J. Watters
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin Madison, Madison, WI 53706, USA; (A.C.E.); (A.O.K.); (S.M.C.S.)
| |
Collapse
|
127
|
Trzeciak P, Herbet M, Dudka J. Common Factors of Alzheimer's Disease and Rheumatoid Arthritis-Pathomechanism and Treatment. Molecules 2021; 26:6038. [PMID: 34641582 PMCID: PMC8512006 DOI: 10.3390/molecules26196038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
The accumulation of amyloid plaques, or misfolded fragments of proteins, leads to the development of a condition known as amyloidosis, which is clinically recognized as a systemic disease. Amyloidosis plays a special role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and rheumatoid arthritis (RA). The occurrence of amyloidosis correlates with the aging process of the organism, and since nowadays, old age is determined by the comfort of functioning and the elimination of unpleasant disease symptoms in the elderly, exposure to this subject is justified. In Alzheimer's disease, amyloid plaques negatively affect glutaminergic and cholinergic transmission and loss of sympathetic protein, while in RA, amyloids stimulated by the activity of the immune system affect the degradation of the osteoarticular bond. The following monograph draws attention to the over-reactivity of the immune system in AD and RA, describes the functionality of the blood-brain barrier as an intermediary medium between RA and AD, and indicates the direction of research to date, focusing on determining the relationship and the cause-effect link between these disorders. The paper presents possible directions for the treatment of amyloidosis, with particular emphasis on innovative therapies.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland; (P.T.)
| | | |
Collapse
|
128
|
Siddiqui A, Shah Z, Jahan RN, Othman I, Kumari Y. Mechanistic role of boswellic acids in Alzheimer's disease: Emphasis on anti-inflammatory properties. Biomed Pharmacother 2021; 144:112250. [PMID: 34607104 DOI: 10.1016/j.biopha.2021.112250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022] Open
Abstract
The resin/gum of Boswellia species belonging to the family of Burseraceae is a naturally occurring mixture of bioactive compounds, which was traditionally used as a folk medicine to treat conditions like chronic inflammation. Several research studies have also explored its' therapeutic potential against multiple neurodegenerative diseases such as Alzheimer's disease (AD). The main chemical constituents of this gum include boswellic acids (BAs) like 3-O-acetyl-11-keto-β boswellic acid (AKBA) that possess potent anti-inflammatory and neuroprotective properties in AD. It is also involved in inhibiting the acetylcholinesterase (AChE) activity in the cholinergic pathway and improve choline levels as well as its binding with nicotinic receptors to produce anti-inflammatory effects. Multiple shreds of evidence have demonstrated that BAs modulate key molecular targets and signalling pathways like 5-lipoxygenase/cyclooxygenase, Nrf2, NF-kB, cholinergic, amyloid-beta (Aβ), and neurofibrillary tangles formation (NFTs) that are involved in AD progression. The present review focuses on the possible mechanistic therapeutic role of BAs in modulating the 5-LOX/COX pathway in arachidonic acid metabolism, activating Nrf2 through binding of ARE, inhibiting NF-kB and AChE activity. In addition, an inhibition of amyloid plaques (Aβ) and neurofibrillary tangles (NFTs) induced neurotoxicity and neuroinflammation in AD by BAs is also discussed in this review. We have also highlighted that BAs possess beneficial effects in AD by targeting multiple molecular pathways and makes it an emerging drug candidate for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Aisha Siddiqui
- Neurological disorder and aging research group (NDA), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Zahoor Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo 43614, OH, USA
| | - Rao Nargis Jahan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Yatinesh Kumari
- Neurological disorder and aging research group (NDA), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia.
| |
Collapse
|
129
|
Coleman LG, Crews FT, Vetreno RP. The persistent impact of adolescent binge alcohol on adult brain structural, cellular, and behavioral pathology: A role for the neuroimmune system and epigenetics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:1-44. [PMID: 34696871 DOI: 10.1016/bs.irn.2021.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adolescence is a critical neurodevelopmental window for maturation of brain structure, neurocircuitry, and glia. This development is sculpted by an individual's unique experiences and genetic background to establish adult level cognitive function and behavioral makeup. Alcohol abuse during adolescence is associated with an increased lifetime risk for developing an alcohol use disorder (AUD). Adolescents participate in heavy, episodic binge drinking that causes persistent changes in neurocircuitry and behavior. These changes may underlie the increased risk for AUD and might also promote cognitive deficits later in life. In this chapter, we have examined research on the persistent effects of adolescent binge-drinking both in humans and in rodent models. These studies implicate roles for neuroimmune signaling as well as epigenetic reprogramming of neurons and glia, which create a vulnerable neuroenvironment. Some of these changes are reversible, giving hope for future treatments to prevent many of the long-term consequences of adolescent alcohol abuse.
Collapse
Affiliation(s)
- Leon G Coleman
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States; Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
130
|
Serrano-Pozo A, Li Z, Noori A, Nguyen HN, Mezlini A, Li L, Hudry E, Jackson RJ, Hyman BT, Das S. Effect of APOE alleles on the glial transcriptome in normal aging and Alzheimer's disease. NATURE AGING 2021; 1:919-931. [PMID: 36199750 PMCID: PMC9531903 DOI: 10.1038/s43587-021-00123-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 09/03/2021] [Indexed: 05/02/2023]
Abstract
The roles of APOEε4 and APOEε2-the strongest genetic risk and protective factors for Alzheimer's disease-in glial responses remain elusive. We tested the hypothesis that APOE alleles differentially impact glial responses by investigating their effects on the glial transcriptome from elderly control brains with no neuritic amyloid plaques. We identified a cluster of microglial genes that are upregulated in APOEε4 and downregulated in APOEε2 carriers relative to APOEε3 homozygotes. This microglia-APOE cluster is enriched in phagocytosis-including TREM2 and TYROBP-and proinflammatory genes, and is also detectable in brains with frequent neuritic plaques. Next, we tested these findings in APOE knock-in mice exposed to acute (lipopolysaccharide challenge) and chronic (cerebral β-amyloidosis) insults and found that these mice partially recapitulate human APOE-linked expression patterns. Thus, the APOEε4 allele might prime microglia towards a phagocytic and proinflammatory state through an APOE-TREM2-TYROBP axis in normal aging as well as in Alzheimer's disease.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Zhaozhi Li
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
| | - Ayush Noori
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
| | - Huong N. Nguyen
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Aziz Mezlini
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Liang Li
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Eloise Hudry
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rosemary J. Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
131
|
Sarrouilhe D, Defamie N, Mesnil M. Is the Exposome Involved in Brain Disorders through the Serotoninergic System? Biomedicines 2021; 9:1351. [PMID: 34680468 PMCID: PMC8533279 DOI: 10.3390/biomedicines9101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 11/24/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a biogenic monoamine acting as a neurotransmitter in the central nervous system (CNS), local mediator in the gut, and vasoactive agent in the blood. It has been linked to a variety of CNS functions and is implicated in many CNS and psychiatric disorders. The high comorbidity between some neuropathies can be partially understood by the fact that these diseases share a common etiology involving the serotoninergic system. In addition to its well-known functions, serotonin has been shown to be a mitogenic factor for a wide range of normal and tumor cells, including glioma cells, in vitro. The developing CNS of fetus and newborn is particularly susceptible to the deleterious effects of neurotoxic substances in our environment, and perinatal exposure could result in the later development of diseases, a hypothesis known as the developmental origin of health and disease. Some of these substances affect the serotoninergic system and could therefore be the source of a silent pandemic of neurodevelopmental toxicity. This review presents the available data that are contributing to the appreciation of the effects of the exposome on the serotoninergic system and their potential link with brain pathologies (neurodevelopmental, neurodegenerative, neurobehavioral disorders, and glioblastoma).
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculté de Médecine et Pharmacie, 6 Rue de la Milétrie, Bât D1, TSA 51115, CEDEX 09, 86073 Poitiers, France
| | - Norah Defamie
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| | - Marc Mesnil
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| |
Collapse
|
132
|
Huang W, Bartosch AM, Xiao H, Maji S, Youth EHH, Flowers X, Leskinen S, Tomljanovic Z, Iodice G, Boyett D, Spinazzi E, Menon V, McGovern RA, McKhann GM, Teich AF. An immune response characterizes early Alzheimer's disease pathology and subjective cognitive impairment in hydrocephalus biopsies. Nat Commun 2021; 12:5659. [PMID: 34580300 PMCID: PMC8476497 DOI: 10.1038/s41467-021-25902-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Early Alzheimer's disease (AD) pathology can be found in cortical biopsies taken during shunt placement for Normal Pressure Hydrocephalus. This represents an opportunity to study early AD pathology in living patients. Here we report RNA-seq data on 106 cortical biopsies from this patient population. A restricted set of genes correlate with AD pathology in these biopsies, and co-expression network analysis demonstrates an evolution from microglial homeostasis to a disease-associated microglial phenotype in conjunction with increasing AD pathologic burden, along with a subset of additional astrocytic and neuronal genes that accompany these changes. Further analysis demonstrates that these correlations are driven by patients that report mild cognitive symptoms, despite similar levels of biopsy β-amyloid and tau pathology in comparison to patients who report no cognitive symptoms. Taken together, these findings highlight a restricted set of microglial and non-microglial genes that correlate with early AD pathology in the setting of subjective cognitive decline.
Collapse
Affiliation(s)
- Wenrui Huang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Anne Marie Bartosch
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Suvrajit Maji
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Elliot H H Youth
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Sandra Leskinen
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Zeljko Tomljanovic
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Gail Iodice
- Department of Neurosurgery, Columbia University, New York, NY, USA
| | - Deborah Boyett
- Department of Neurosurgery, Columbia University, New York, NY, USA
| | | | - Vilas Menon
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | - Robert A McGovern
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Guy M McKhann
- Department of Neurosurgery, Columbia University, New York, NY, USA
| | - Andrew F Teich
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- Department of Neurology, Columbia University, New York, NY, USA.
| |
Collapse
|
133
|
Correia SC, Moreira PI. Oxygen Sensing and Signaling in Alzheimer's Disease: A Breathtaking Story! Cell Mol Neurobiol 2021; 42:3-21. [PMID: 34510330 DOI: 10.1007/s10571-021-01148-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Oxygen sensing and homeostasis is indispensable for the maintenance of brain structural and functional integrity. Under low-oxygen tension, the non-diseased brain has the ability to cope with hypoxia by triggering a homeostatic response governed by the highly conserved hypoxia-inducible family (HIF) of transcription factors. With the advent of advanced neuroimaging tools, it is now recognized that cerebral hypoperfusion, and consequently hypoxia, is a consistent feature along the Alzheimer's disease (AD) continuum. Of note, the reduction in cerebral blood flow and tissue oxygenation detected during the prodromal phases of AD, drastically aggravates as disease progresses. Within this scenario a fundamental question arises: How HIF-driven homeostatic brain response to hypoxia "behaves" during the AD continuum? In this sense, the present review is aimed to critically discuss and summarize the current knowledge regarding the involvement of hypoxia and HIF signaling in the onset and progression of AD pathology. Importantly, the promises and challenges of non-pharmacological and pharmacological strategies aimed to target hypoxia will be discussed as a new "hope" to prevent and/or postpone the neurodegenerative events that occur in the AD brain.
Collapse
Affiliation(s)
- Sónia C Correia
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal. .,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
134
|
Boström G, Freyhult E, Virhammar J, Alcolea D, Tumani H, Otto M, Brundin RM, Kilander L, Löwenmark M, Giedraitis V, Lleó A, von Arnim CAF, Kultima K, Ingelsson M. Different Inflammatory Signatures in Alzheimer's Disease and Frontotemporal Dementia Cerebrospinal Fluid. J Alzheimers Dis 2021; 81:629-640. [PMID: 33814444 PMCID: PMC8203220 DOI: 10.3233/jad-201565] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Neuroinflammatory processes are common in neurodegenerative diseases such as Alzheimer’s disease (AD) and frontotemporal dementia (FTD), but current knowledge is limited as to whether cerebrospinal fluid (CSF) levels of neuroinflammatory proteins are altered in these diseases. Objective: To identify and characterize neuroinflammatory signatures in CSF from patients with AD, mild cognitive impairment (MCI), and FTD. Methods: We used proximity extension assay and ANOVA to measure and compare levels of 92 inflammatory proteins in CSF from 42 patients with AD, 29 with MCI due to AD (MCI/AD), 22 with stable MCI, 42 with FTD, and 49 control subjects, correcting for age, gender, collection unit, and multiple testing. Results: Levels of matrix metalloproteinase-10 (MMP-10) were increased in AD, MCI/AD, and FTD compared with controls (AD: fold change [FC] = 1.32, 95% confidence interval [CI] 1.14–1.53, q = 0.018; MCI/AD: FC = 1.53, 95% CI 1.20–1.94, q = 0.045; and FTD: FC = 1.42, 95% CI 1.10–1.83, q = 0.020). MMP-10 and eleven additional proteins were increased in MCI/AD, compared with MCI (q < 0.05). In FTD, 36 proteins were decreased, while none was decreased in AD or MCI/AD, compared with controls (q < 0.05). Conclusion: In this cross-sectional multi-center study, we found distinct patterns of CSF inflammatory marker levels in FTD and in both early and established AD, suggesting differing neuroinflammatory processes in the two disorders.
Collapse
Affiliation(s)
- Gustaf Boström
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Eva Freyhult
- Department of Medical Sciences, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Virhammar
- Department of Neuroscience, Neurology, Uppsala University Hospital, Uppsala, Sweden
| | - Daniel Alcolea
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Rose-Marie Brundin
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Malin Löwenmark
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Alberto Lleó
- Memory Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Christine A F von Arnim
- Department of Neurology, Ulm University Hospital, Ulm, Germany.,Department of Geriatric Medicine, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
135
|
Yin X, Qiu Y, Zhao C, Zhou Z, Bao J, Qian W. The Role of Amyloid-Beta and Tau in the Early Pathogenesis of Alzheimer's Disease. Med Sci Monit 2021; 27:e933084. [PMID: 34471085 PMCID: PMC8422899 DOI: 10.12659/msm.933084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The abnormal accumulation of amyloid-b (Ab) and neurofibrillary tangles (NFTs) containing phosphorylated tau proteins are the main histopathological feature of Alzheimer's disease (AD). Synaptic damage and loss are earlier events than amyloid plaques and NFTs in AD progress and best correlate with cognitive deficits in AD patients. Soluble oligomeric Aß initiates the progression of AD and tau mediates the subsequent synaptic impairments at an early stage of AD. In this review we discuss how Ab or/and tau causes synaptic dysfunction. Ab oligomers gather at synapses and give rise to synaptic death in a variety of ways such as regulating receptors and receptor tyrosine kinases, unbalancing calcium homeostasis, and activating caspases and calcineurin. A large amount of hyperphosphorylated tau exists in the synapse of the AD brain. Aß-triggered synaptic deficits are dependent on tau. Soluble, hyperphosphorylated tau is much more correlated to cognitive decline in AD patients. Tau-targeted therapies have received more attention because the treatments targeting Aß failed in AD. Here, we also review the therapy strategies used to intervene in the very early stages of AD. Soluble hyperphosphorylated tau forms a complex with cell surface receptors, scaffold proteins, or intracellular signaling molecules to damage synaptic function. Therefore, therapeutic strategies targeting synaptic tau at the early stage of AD may ameliorating pathology in AD. This review aims to provide an update on the role of oligomeric Ab and soluble hyperphosphorylated tau in the early pathogenesis of Alzheimer's disease and to develop a new treatment strategy based on this.
Collapse
Affiliation(s)
- Xiaomin Yin
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China (mainland).,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Yanyan Qiu
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Chenhao Zhao
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Zheng Zhou
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Junze Bao
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Wei Qian
- Department of Biochemistry and Molecular Biology, Medical School, Nantong University, Nantong, Jiangsu, China (mainland).,Jiangsu Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China (mainland).,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China (mainland)
| |
Collapse
|
136
|
Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, Savard M, Lussier FZ, Tissot C, Karikari TK, Ottoy J, Mathotaarachchi S, Stevenson J, Massarweh G, Schöll M, de Leon MJ, Soucy JP, Edison P, Blennow K, Zetterberg H, Gauthier S, Rosa-Neto P. Microglial activation and tau propagate jointly across Braak stages. Nat Med 2021; 27:1592-1599. [PMID: 34446931 DOI: 10.1038/s41591-021-01456-w] [Citation(s) in RCA: 240] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/28/2021] [Indexed: 11/09/2022]
Abstract
Compelling experimental evidence suggests that microglial activation is involved in the spread of tau tangles over the neocortex in Alzheimer's disease (AD). We tested the hypothesis that the spatial propagation of microglial activation and tau accumulation colocalize in a Braak-like pattern in the living human brain. We studied 130 individuals across the aging and AD clinical spectrum with positron emission tomography brain imaging for microglial activation ([11C]PBR28), amyloid-β (Aβ) ([18F]AZD4694) and tau ([18F]MK-6240) pathologies. We further assessed microglial triggering receptor expressed on myeloid cells 2 (TREM2) cerebrospinal fluid (CSF) concentrations and brain gene expression patterns. We found that [11C]PBR28 correlated with CSF soluble TREM2 and showed regional distribution resembling TREM2 gene expression. Network analysis revealed that microglial activation and tau correlated hierarchically with each other following Braak-like stages. Regression analysis revealed that the longitudinal tau propagation pathways depended on the baseline microglia network rather than the tau network circuits. The co-occurrence of Aβ, tau and microglia abnormalities was the strongest predictor of cognitive impairment in our study population. Our findings support a model where an interaction between Aβ and activated microglia sets the pace for tau spread across Braak stages.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Departments of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada. .,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - Andrea L Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley, NHS Foundation, London, UK
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julie Ottoy
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium.,LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Gassan Massarweh
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Michael Schöll
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Mony J de Leon
- Department of Radiology Weill Medical Center Brain Health Imaging Institute, Cornell University, Ithaca, NY, USA
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada. .,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
137
|
Vasilopoulou F, Escolano C, Pallàs M, Griñán-Ferré C. Microarray Analysis Revealed Inflammatory Transcriptomic Changes after LSL60101 Treatment in 5XFAD Mice Model. Genes (Basel) 2021; 12:1315. [PMID: 34573297 PMCID: PMC8468036 DOI: 10.3390/genes12091315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022] Open
Abstract
I2-IR have been found dysregulated in patients with neurodegenerative diseases, such as Alzheimer's disease (AD), in which the importance of neuroinflammation in the establishment and maintenance of cognitive decline is well-documented. To research the implication of I2-IR in neuroinflammatory pathways altered in AD, we determined the expression profile of genes associated with inflammation in the 5XFAD model treated with LSL60101, a well-established I2-IR ligand. Thus, we performed a qPCR array containing 84 inflammation-related genes. Hierarchical clustering analysis revealed three gene clusters, suggesting that treatment with LSL60101 affects the gene expression associated with inflammation in the 5XFAD model. Furthermore, we evaluated the functions of the three clusters; thereby performing a pathway enrichment analysis using the GO database. As we expected, clusters 2 and 3 showed alterations in the inflammatory response, chemotaxis and the chemokine-mediated signaling pathway, among others. To validate previous results from the gene profiling analysis, the expression levels of a representative subset of mRNAs were selected according to the intensity of the observed changes and their biological relevance. Interestingly, changes induced by LSL60101 in the 5XFAD model were validated for several genes. These results suggest that treatment with LSL60101 in the 5XFAD model reverses the inflammatory process during the development of AD.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain;
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| |
Collapse
|
138
|
Buccellato FR, D’Anca M, Fenoglio C, Scarpini E, Galimberti D. Role of Oxidative Damage in Alzheimer's Disease and Neurodegeneration: From Pathogenic Mechanisms to Biomarker Discovery. Antioxidants (Basel) 2021; 10:antiox10091353. [PMID: 34572985 PMCID: PMC8471953 DOI: 10.3390/antiox10091353] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder accounting for over 50% of all dementia patients and representing a leading cause of death worldwide for the global ageing population. The lack of effective treatments for overt AD urges the discovery of biomarkers for early diagnosis, i.e., in subjects with mild cognitive impairment (MCI) or prodromal AD. The brain is exposed to oxidative stress as levels of reactive oxygen species (ROS) are increased, whereas cellular antioxidant defenses are decreased. Increased ROS levels can damage cellular structures or molecules, leading to protein, lipid, DNA, or RNA oxidation. Oxidative damage is involved in the molecular mechanisms which link the accumulation of amyloid-β and neurofibrillary tangles, containing hyperphosphorylated tau, to microglia response. In this scenario, microglia are thought to play a crucial role not only in the early events of AD pathogenesis but also in the progression of the disease. This review will focus on oxidative damage products as possible peripheral biomarkers in AD and in the preclinical phases of the disease. Particular attention will be paid to biological fluids such as blood, CSF, urine, and saliva, and potential future use of molecules contained in such body fluids for early differential diagnosis and monitoring the disease course. We will also review the role of oxidative damage and microglia in the pathogenesis of AD and, more broadly, in neurodegeneration.
Collapse
Affiliation(s)
- Francesca Romana Buccellato
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Correspondence: ; Tel.: +39-02 55033814
| | - Marianna D’Anca
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
| | - Elio Scarpini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| |
Collapse
|
139
|
Blood-Based Biomarkers of Neuroinflammation in Alzheimer's Disease: A Central Role for Periphery? Diagnostics (Basel) 2021; 11:diagnostics11091525. [PMID: 34573867 PMCID: PMC8464786 DOI: 10.3390/diagnostics11091525] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation represents a central feature in the development of Alzheimer’s disease (AD). The resident innate immune cells of the brain are the principal players in neuroinflammation, and their activation leads to a defensive response aimed at promoting β-amyloid (Aβ) clearance. However, it is now widely accepted that the peripheral immune system—by virtue of a dysfunctional blood–brain barrier (BBB)—is involved in the pathogenesis and progression of AD; microglial and astrocytic activation leads to the release of chemokines able to recruit peripheral immune cells into the central nervous system (CNS); at the same time, cytokines released by peripheral cells are able to cross the BBB and act upon glial cells, modifying their phenotype. To successfully fight this neurodegenerative disorder, accurate and sensitive biomarkers are required to be used for implementing an early diagnosis, monitoring the disease progression and treatment effectiveness. Interestingly, as a result of the bidirectional communication between the brain and the periphery, the blood compartment ends up reflecting several pathological changes occurring in the AD brain and can represent an accessible source for such biomarkers. In this review, we provide an overview on some of the most promising peripheral biomarkers of neuroinflammation, discussing their pathogenic role in AD.
Collapse
|
140
|
Highlighting Immune System and Stress in Major Depressive Disorder, Parkinson's, and Alzheimer's Diseases, with a Connection with Serotonin. Int J Mol Sci 2021; 22:ijms22168525. [PMID: 34445231 PMCID: PMC8395198 DOI: 10.3390/ijms22168525] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 01/10/2023] Open
Abstract
There is recognition that both stress and immune responses are important factors in a variety of neurological disorders. Moreover, there is an important role of several neurotransmitters that connect these factors to several neurological diseases, with a special focus in this paper on serotonin. Accordingly, it is known that imbalances in stressors can promote a variety of neuropsychiatric or neurodegenerative pathologies. Here, we discuss some facts that link major depressive disorder, Alzheimer’s, and Parkinson’s to the stress and immune responses, as well as the connection between these responses and serotonergic signaling. These are important topics of investigation which may lead to new or better treatments, improving the life quality of patients that suffer from these conditions.
Collapse
|
141
|
Viejo L, Noori A, Merrill E, Das S, Hyman BT, Serrano-Pozo A. Systematic review of human post-mortem immunohistochemical studies and bioinformatics analyses unveil the complexity of astrocyte reaction in Alzheimer's disease. Neuropathol Appl Neurobiol 2021; 48:e12753. [PMID: 34297416 PMCID: PMC8766893 DOI: 10.1111/nan.12753] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
AIMS Reactive astrocytes in Alzheimer's disease (AD) have traditionally been demonstrated by increased glial fibrillary acidic protein (GFAP) immunoreactivity; however, astrocyte reaction is a complex and heterogeneous phenomenon involving multiple astrocyte functions beyond cytoskeletal remodelling. To better understand astrocyte reaction in AD, we conducted a systematic review of astrocyte immunohistochemical studies in post-mortem AD brains followed by bioinformatics analyses on the extracted reactive astrocyte markers. METHODS NCBI PubMed, APA PsycInfo and WoS-SCIE databases were interrogated for original English research articles with the search terms 'Alzheimer's disease' AND 'astrocytes.' Bioinformatics analyses included protein-protein interaction network analysis, pathway enrichment, and transcription factor enrichment, as well as comparison with public human -omics datasets. RESULTS A total of 306 articles meeting eligibility criteria rendered 196 proteins, most of which were reported to be upregulated in AD vs control brains. Besides cytoskeletal remodelling (e.g., GFAP), bioinformatics analyses revealed a wide range of functional alterations including neuroinflammation (e.g., IL6, MAPK1/3/8 and TNF), oxidative stress and antioxidant defence (e.g., MT1A/2A, NFE2L2, NOS1/2/3, PRDX6 and SOD1/2), lipid metabolism (e.g., APOE, CLU and LRP1), proteostasis (e.g., cathepsins, CRYAB and HSPB1/2/6/8), extracellular matrix organisation (e.g., CD44, MMP1/3 and SERPINA3), and neurotransmission (e.g., CHRNA7, GABA, GLUL, GRM5, MAOB and SLC1A2), among others. CTCF and ESR1 emerged as potential transcription factors driving these changes. Comparison with published -omics datasets validated our results, demonstrating a significant overlap with reported transcriptomic and proteomic changes in AD brains and/or CSF. CONCLUSIONS Our systematic review of the neuropathological literature reveals the complexity of AD reactive astrogliosis. We have shared these findings as an online resource available at www.astrocyteatlas.org.
Collapse
Affiliation(s)
- Lucía Viejo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ayush Noori
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Harvard College, Cambridge, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA
| | - Emily Merrill
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
142
|
Thomsen BB, Madsen C, Krohn KT, Thygesen C, Schütt T, Metaxas A, Darvesh S, Agerholm JS, Wirenfeldt M, Berendt M, Finsen B. Mild Microglial Responses in the Cortex and Perivascular Macrophage Infiltration in Subcortical White Matter in Dogs with Age-Related Dementia Modelling Prodromal Alzheimer's Disease. J Alzheimers Dis 2021; 82:575-592. [PMID: 34057083 PMCID: PMC8385501 DOI: 10.3233/jad-210040] [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] [Indexed: 12/15/2022]
Abstract
Background: Microglia contribute to Alzheimer’s disease (AD) pathogenesis by clearing amyloid-β (Aβ) and driving neuroinflammation. Domestic dogs with age-related dementia (canine cognitive dysfunction (CCD)) develop cerebral amyloidosis like humans developing AD, and studying such dogs can provide novel information about microglial response in prodromal AD. Objective: The aim was to investigate the microglial response in the cortical grey and the subcortical white matter in dogs with CCD versus age-matched cognitively normal dogs. Methods: Brains from aged dogs with CCD and age-matched controls without dementia were studied. Cases were defined by dementia rating score. Brain sections were stained for Aβ, thioflavin S, hyperphosphorylated tau, and the microglial-macrophage ionized calcium binding adaptor molecule 1 (Iba1). Results were correlated to dementia rating score and tissue levels of Aβ. Results: Microglial numbers were higher in the Aβ plaque-loaded deep cortical layers in CCD versus control dogs, while the coverage by microglial processes were comparable. Aβ plaques were of the diffuse type and without microglial aggregation. However, a correlation was found between the %Iba1 area and insoluble Aβ 42 and N-terminal pyroglutamate modified Aβ(N3pE)-42. The %Iba1 area was higher in white matter, showing phosphorylation of S396 tau, versus grey matter. Perivascular macrophage infiltrates were abundant in the white matter particularly in CDD dogs. Conclusion: The results from this study of the microglial-macrophage response in dogs with CCD are suggestive of relatively mild microglial responses in the Aβ plaque-loaded deep cortical layers and perivascular macrophage infiltrates in the subcortical white matter, in prodromal AD.
Collapse
Affiliation(s)
- Barbara Blicher Thomsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Madsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Katrine Tækker Krohn
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Camilla Thygesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Trine Schütt
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Athanasios Metaxas
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,School of Science, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Division of Neurology and Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jørgen Steen Agerholm
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Wirenfeldt
- BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Institute of Clinical Science, Odense University Hospital, Odense, Denmark
| | - Mette Berendt
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
143
|
Wegiel J, Flory M, Kuchna I, Nowicki K, Ma SY, Wegiel J, Badmaev E, Leon MD, Wisniewski T, Reisberg B. Clinicopathological Staging of Dynamics of Neurodegeneration and Neuronal Loss in Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:21-44. [PMID: 33270870 DOI: 10.1093/jnen/nlaa140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clinical and neuropathological staging of Alzheimer disease (AD) neurodegeneration and neuronal loss dynamics is the baseline for identification of treatment targets and timing. The aim of this study of 14 brain regions in 25 subjects diagnosed with AD and 13 age-matched control subjects was to establish the pattern of neurodegeneration, and the severity and rate of neuronal loss in mild cognitive impairment/mild AD (Functional Assessment Staging [FAST] test 3-4), moderate to moderately severe AD (FAST 5-6), and severe AD (FAST 7). The study revealed (1) the most severe neuronal loss in FAST 3-4; (2) the highest rate of neuronal loss in FAST 5-6, to the "critical" point limiting further increase in neuronal loss; (3) progression of neurofibrillary degeneration, but decline of neuronal loss to a floor level in FAST 7; and (4) structure-specific rate of neuronal loss caused by neurofibrillary degeneration and a large pool of neuronal loss caused by other mechanisms. This study defines a range and speed of progression of AD pathology and functional decline that might potentially be prevented by the arrest of neuronal loss, both related and unrelated to neurofibrillary degeneration, during the 9-year duration of mild cognitive impairment/mild AD.
Collapse
Affiliation(s)
| | - Michael Flory
- Research Design and Analysis Service, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | | | | | | | - Jarek Wegiel
- From the Department of Developmental Neurobiology
| | | | | | - Thomas Wisniewski
- Departments of Neurology, Pathology, and Psychiatry, NYU Langone Medical Center, New York, New York
| | - Barry Reisberg
- Departments of Neurology, Pathology, and Psychiatry, NYU Langone Medical Center, New York, New York
| |
Collapse
|
144
|
Bellaver B, Ferrari-Souza JP, Uglione da Ros L, Carter SF, Rodriguez-Vieitez E, Nordberg A, Pellerin L, Rosa-Neto P, Leffa DT, Zimmer ER. Astrocyte Biomarkers in Alzheimer Disease: A Systematic Review and Meta-analysis. Neurology 2021; 96:e2944-e2955. [PMID: 33952650 DOI: 10.1212/wnl.0000000000012109] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/19/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to determine whether fluid and imaging astrocyte biomarkers are altered in Alzheimer disease (AD). METHODS PubMed and Web of Science databases were searched for articles reporting fluid or imaging astrocyte biomarkers in AD. Pooled effect sizes were determined with standardized mean differences (SMDs) using the Hedge G method with random effects to determine biomarker performance. Adapted questions from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42020192304). RESULTS The initial search identified 1,425 articles. After exclusion criteria were applied, 33 articles (a total of 3,204 individuals) measuring levels of glial fibrillary acidic protein (GFAP), S100B, chitinase-3-like protein 1 (YKL-40), and aquaporin 4 in the blood and CSF, as well as monoamine oxidase-B indexed by PET 11C-deuterium-l-deprenyl, were included. GFAP (SMD 0.94, 95% confidence interval [CI] 0.71-1.18) and YKL-40 (SMD 0.76, 95% CI 0.63-0.89) levels in the CSF and S100B levels in the blood (SMD 2.91, 95% CI 1.01-4.8) were found to be significantly increased in patients with AD. CONCLUSIONS Despite significant progress, applications of astrocyte biomarkers in AD remain in their early days. This meta-analysis demonstrated that astrocyte biomarkers are consistently altered in AD and supports further investigation for their inclusion in the AD clinical research framework for observational and interventional studies.
Collapse
Affiliation(s)
- Bruna Bellaver
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Lucas Uglione da Ros
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Stephen F Carter
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Elena Rodriguez-Vieitez
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Agneta Nordberg
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Luc Pellerin
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Douglas Teixeira Leffa
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil.
| |
Collapse
|
145
|
Liang Y, Raven F, Ward JF, Zhen S, Zhang S, Sun H, Miller SJ, Choi SH, Tanzi RE, Zhang C. Upregulation of Alzheimer's Disease Amyloid-β Protein Precursor in Astrocytes Both in vitro and in vivo. J Alzheimers Dis 2021; 76:1071-1082. [PMID: 32597805 DOI: 10.3233/jad-200128] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The amyloid cascade hypothesis of Alzheimer's disease (AD) posits that amyloid-β (Aβ) protein accumulation underlies the pathogenesis of the disease by leading to the formation of amyloid plaques, a pathologic hallmark of AD. Aβ is a proteolytic product of amyloid-β protein precursor (AβPP; APP), which is expressed in both neurons and astrocytes. Although considerable evidence shows that astrocytes may play critical roles in the pathogenesis of AD, the longitudinal changes of amyloid plaques in relationship to AβPP expression in astrocytes and cellular consequences are largely unknown. OBJECTIVE Here, we aimed to investigate astrocyte-related pathological changes of Aβ and AβPP using immunohistochemistry and biochemical studies in both animal and cell models. METHODS/RESULTS We utilized 5XFAD transgenic mice and found age-dependent upregulation of AβPP in astrocytes demonstrated with astrocytic reactive properties, which followed appearance of amyloid plaques in the brain. We also observed that AβPP proteins presented well-defined punctate immuno reactivity in young animals, whereas AβPP staining showed disrupted structures surrounding amyloid plaques in older mice. Moreover, we utilized astrocyte cell models and showed that pretreatment of Aβ42 resulted in downstream astrocyte autonomous changes, including up regulation in AβPP and BACE1 levels, as well as prolonged amyloidogenesis that could be reduced by pharmacological inhibition of BACE1. CONCLUSION Collectively, our results show that age-dependent AβPP up regulation in astrocytes is a key feature in AD, which will not only provide novel insights for understanding AD progression, but also may offer new therapeutic strategies for treating AD.
Collapse
Affiliation(s)
- Yingxia Liang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Frank Raven
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Joseph F Ward
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sherri Zhen
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Siyi Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Haoqi Sun
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Sean J Miller
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
146
|
Ji Z, Liu C, Zhao W, Soto C, Zhou X. Multi-scale modeling for systematically understanding the key roles of microglia in AD development. Comput Biol Med 2021; 133:104374. [PMID: 33864975 DOI: 10.1016/j.compbiomed.2021.104374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of age-related dementia, affecting over 5 million people in the United States. Unfortunately, current therapies are largely palliative and several potential drug candidates have failed in late-stage clinical trials. Studies suggest that microglia-mediated neuroinflammation might be responsible for the failures of various therapies. Microglia contribute to Aβ clearance in the early stage of neurodegeneration and may contribute to AD development at the late stage by releasing pro-inflammatory cytokines. However, the activation profile and phenotypic changes of microglia during the development of AD are poorly understood. To systematically understand the key role of microglia in AD progression and predict the optimal therapeutic strategy in silico, we developed a 3D multi-scale model of AD (MSMAD) by integrating multi-level experimental data, to manipulate the neurodegeneration in a simulated system. Based on our analysis, we revealed that how TREM2-related signal transduction leads to an imbalance in the activation of different microglia phenotypes, thereby promoting AD development. Our MSMAD model also provides an optimal therapeutic strategy for improving the outcome of AD treatment.
Collapse
Affiliation(s)
- Zhiwei Ji
- College of Artificial Intelligence, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, Jiangsu, 210095, China; School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA.
| | - Changan Liu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Weiling Zhao
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease & Brain Disorder, Department of Neurology, The University of Texas McGovern Medical School, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Xiaobo Zhou
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
147
|
Edler MK, Mhatre-Winters I, Richardson JR. Microglia in Aging and Alzheimer's Disease: A Comparative Species Review. Cells 2021; 10:1138. [PMID: 34066847 PMCID: PMC8150617 DOI: 10.3390/cells10051138] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system that help nourish and support neurons, clear debris, and respond to foreign stimuli. Greatly impacted by their environment, microglia go through rapid changes in cell shape, gene expression, and functional behavior during states of infection, trauma, and neurodegeneration. Aging also has a profound effect on microglia, leading to chronic inflammation and an increase in the brain's susceptibility to neurodegenerative processes that occur in Alzheimer's disease. Despite the scientific community's growing knowledge in the field of neuroinflammation, the overall success rate of drug treatment for age-related and neurodegenerative diseases remains incredibly low. Potential reasons for the lack of translation from animal models to the clinic include the use of a single species model, an assumption of similarity in humans, and ignoring contradictory data or information from other species. To aid in the selection of validated and predictive animal models and to bridge the translational gap, this review evaluates similarities and differences among species in microglial activation and density, morphology and phenotype, cytokine expression, phagocytosis, and production of oxidative species in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44240, USA;
| | - Isha Mhatre-Winters
- School of Biomedical Sciences, College of Arts and Sciences, Kent State University, Kent, OH 44240, USA;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jason R. Richardson
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
148
|
Neuroinflammation in Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9050524. [PMID: 34067173 PMCID: PMC8150909 DOI: 10.3390/biomedicines9050524] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease associated with human aging. Ten percent of individuals over 65 years have AD and its prevalence continues to rise with increasing age. There are currently no effective disease modifying treatments for AD, resulting in increasingly large socioeconomic and personal costs. Increasing age is associated with an increase in low-grade chronic inflammation (inflammaging) that may contribute to the neurodegenerative process in AD. Although the exact mechanisms remain unclear, aberrant elevation of reactive oxygen and nitrogen species (RONS) levels from several endogenous and exogenous processes in the brain may not only affect cell signaling, but also trigger cellular senescence, inflammation, and pyroptosis. Moreover, a compromised immune privilege of the brain that allows the infiltration of peripheral immune cells and infectious agents may play a role. Additionally, meta-inflammation as well as gut microbiota dysbiosis may drive the neuroinflammatory process. Considering that inflammatory/immune pathways are dysregulated in parallel with cognitive dysfunction in AD, elucidating the relationship between the central nervous system and the immune system may facilitate the development of a safe and effective therapy for AD. We discuss some current ideas on processes in inflammaging that appear to drive the neurodegenerative process in AD and summarize details on a few immunomodulatory strategies being developed to selectively target the detrimental aspects of neuroinflammation without affecting defense mechanisms against pathogens and tissue damage.
Collapse
|
149
|
Kanaan NM, Grabinski T. Neuronal and Glial Distribution of Tau Protein in the Adult Rat and Monkey. Front Mol Neurosci 2021; 14:607303. [PMID: 33986642 PMCID: PMC8112591 DOI: 10.3389/fnmol.2021.607303] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/23/2021] [Indexed: 12/27/2022] Open
Abstract
Tau is a microtubule-associated protein for which the physiological functions remain a topic of vigorous investigation. Additionally, tau is a central player in the pathogenesis of several diseases such as Alzheimer's disease and several frontotemporal dementias. A critical variable to understanding tau in physiological and disease contexts is its normal localization within cells of the adult CNS. Tau is often described as an axon-specific (or enriched) and neuron-specific protein with little to no expression in glial cells, all of which are untrue. Understanding normal tau distribution also impacts interpretation of experimental results and hypotheses regarding its role in disease. Thus, we set out to help clarify the normal localization of tau in the adult CNS of middle-aged rats and rhesus macaque using the hippocampus as a representative brain structure. The physiological concentration of tau in the rat hippocampus was 6.6 μM and in white matter was 3.6 μM as determined by quantitative sandwich ELISAs. We evaluated the cellular localization of tau using multiple tau-specific antibodies with epitopes to different regions, including Tau1, Tau5, Tau7, R1, and two novel primate-specific antibodies NT9 and NT15. In the rat and monkey, tau was localized within the somatodendritic and axonal compartments, as well as a subset of neuronal nuclei. Semi-quantitative fluorescence intensity measurements revealed that depending on the specific reagent used the somatodendritic tau is relatively equal to, higher than, or lower than axonal tau, highlighting differential labeling of tau with various antibodies despite its distribution throughout the neuron. Tau was strongly expressed in mature oligodendrocytes and displayed little to no expression in oligodendrocyte precursor cells, astrocytes or microglia. Collectively, the data indicate tau is ∼3 - 7 μM under physiological conditions, is not specifically enriched in axons, and is normally found in both neurons and mature oligodendrocytes in the adult CNS. The full landscape of tau distribution is not revealed by all antibodies suggesting availability of the epitopes is different within specific neuronal compartments. These findings set the stage for better understanding normal tau distributions and interpreting data regarding the presence of tau in different compartments or cell types within disease conditions.
Collapse
Affiliation(s)
- Nicholas M. Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Neuroscience Program, Michigan State University, East Lansing, MI, United States
- Mercy Health Hauenstein Neuroscience Center, Grand Rapids, MI, United States
| | - Tessa Grabinski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| |
Collapse
|
150
|
Uddin MS, Kabir MT, Al Mamun A, Behl T, Mansouri RA, Aloqbi AA, Perveen A, Hafeez A, Ashraf GM. Exploring Potential of Alkaloidal Phytochemicals Targeting Neuroinflammatory Signaling of Alzheimer's Disease. Curr Pharm Des 2021; 27:357-366. [PMID: 32473620 DOI: 10.2174/1381612826666200531151004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is marked by cognitive dysfunctions and the existence of neuropathological hallmarks such as amyloid plaques, and neurofibrillary tangles. It has been observed that a persistent immune response in the brain has appeared as another neuropathological hallmark in AD. The sustained activation of the microglia, the brain's resident macrophages, and other immune cells has been shown to aggravate both tau and amyloid pathology and may consider as a connection in the AD pathogenesis. However, the basic mechanisms that link immune responses in the pathogenesis of AD are unclear until now since the process of neuroinflammation can have either a harmful or favorable effect on AD, according to the phase of the disease. Numerous researches recommend that nutritional fruits, as well as vegetables, possess neurodefensive properties against the detrimental effects of neuroinflammation and aging. Moreover, these effects are controlled by diverse phytochemical compounds that are found in plants and demonstrate anti-inflammatory, neuroprotective, as well as other beneficial actions. In this review, we focus on the link of neuroinflammation in AD as well as highlight the probable mechanisms of alkaloidal phytochemicals to combat the neuroinflammatory aspect of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rasha A Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|