493 results on '"Boche, Delphine"'
Search Results
152. Long-term effects of Aβ42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial
- Author
-
Holmes, Clive, primary, Boche, Delphine, additional, Wilkinson, David, additional, Yadegarfar, Ghasem, additional, Hopkins, Vivienne, additional, Bayer, Anthony, additional, Jones, Roy W, additional, Bullock, Roger, additional, Love, Seth, additional, Neal, James W, additional, Zotova, Elina, additional, and Nicoll, James AR, additional
- Published
- 2008
- Full Text
- View/download PDF
153. SYMPOSIUM: Clearance of Aβ from the Brain in Alzheimer' Disease: The Role of the Immune System in Clearance of Aβ from the Brain
- Author
-
Boche, Delphine, primary and Nicoll, James A.R., additional
- Published
- 2008
- Full Text
- View/download PDF
154. Immunotherapy for Alzheimer's Disease and Other Dementias
- Author
-
Boche, Delphine, primary, Nicoll, James A.R., additional, and Weller, Roy O., additional
- Published
- 2006
- Full Text
- View/download PDF
155. Immunotherapy for Alzheimer's disease and other dementias
- Author
-
Boche, Delphine, primary, Nicoll, James AR, additional, and Weller, Roy O, additional
- Published
- 2005
- Full Text
- View/download PDF
156. Comparison of Inflammatory and Acute-Phase Responses in the Brain and Peripheral Organs of the ME7 Model of Prion Disease
- Author
-
Cunningham, Colm, primary, Wilcockson, David C., additional, Boche, Delphine, additional, and Perry, V. Hugh, additional
- Published
- 2005
- Full Text
- View/download PDF
157. Mannose receptor expression specifically reveals perivascular macrophages in normal, injured, and diseased mouse brain
- Author
-
Galea, Ian, primary, Palin, Karine, additional, Newman, Tracey A., additional, Van Rooijen, Nico, additional, Perry, V. Hugh, additional, and Boche, Delphine, additional
- Published
- 2004
- Full Text
- View/download PDF
158. Transforming Growth Factor-β1-Mediated Neuroprotection against Excitotoxic Injuryin Vivo
- Author
-
Boche, Delphine, primary, Cunningham, Colm, additional, Gauldie, Jack, additional, and Perry, V Hugh, additional
- Published
- 2003
- Full Text
- View/download PDF
159. Atypical inflammation in the central nervous system in prion disease
- Author
-
Perry, Hugh V., primary, Cunningham, Colm, additional, and Boche, Delphine, additional
- Published
- 2002
- Full Text
- View/download PDF
160. Aβ immunotherapy for Alzheimer's disease: effects on apoE and cerebral vasculopathy.
- Author
-
Sakai, Kenji, Boche, Delphine, Carare, Roxana, Johnston, David, Holmes, Clive, Love, Seth, and Nicoll, James
- Subjects
IMMUNOTHERAPY ,ALZHEIMER'S disease treatment ,CEREBRAL amyloid angiopathy ,APOLIPOPROTEINS ,IMMUNOHISTOCHEMISTRY - Abstract
Aβ immunotherapy for Alzheimer's disease (AD) results in the removal of Aβ plaques and increased cerebral amyloid angiopathy (CAA). In current clinical trials, amyloid-related imaging abnormalities (ARIAs), putatively due to exacerbation of CAA, are concerning side effects. We aimed to assess the role of the Aβ transporter apolipoprotein E (apoE) in the exacerbation of CAA and development of CAA-associated vasculopathy after Aβ immunotherapy. 12 Aβ42-immunized AD (iAD; AN1792, Elan Pharmaceuticals) cases were compared with 28 unimmunized AD (cAD) cases. Immunohistochemistry was quantified for Aβ42, apoE, apoE E4 and smooth muscle actin, and CAA-associated vasculopathy was analyzed. Aβ immunotherapy was associated with redistribution of apoE from cortical plaques to cerebral vessel walls, mirroring the altered distribution of Aβ42. Concentric vessel wall splitting was increased threefold in leptomeningeal vessels after immunotherapy (cAD 6.3 vs iAD 20.6 %, P < 0.001), but smooth muscle cell abnormalities did not differ. The findings suggest that apoE is involved in the removal of plaques and transport of Aβ to the cerebral vasculature induced by Aβ immunotherapy. Immunotherapy was not associated with CAA-related vascular smooth muscle damage, but was accompanied by increased splitting of the vessel wall, perhaps reflecting enhanced deposition and subsequent removal of Aβ. ARIA occurring in some current trials of Aβ immunotherapy may reflect an extreme form of these vascular changes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
161. Viral load and neuropathology in the SIV model
- Author
-
Boche, Delphine, primary, Khatissian, Emmanuel, additional, Gray, Françoise, additional, Falanga, Pierre, additional, Montagnier, Luc, additional, and Hurtrel, Bruno, additional
- Published
- 1999
- Full Text
- View/download PDF
162. Characterizing inflammatory components in human Alzheimer's disease and following active beta-amyloid 42 immunization
- Author
-
Boche, Delphine, Zotova, Elina, Bharambe, Viraj, Cheaveau, Matthew, Morgan, William, Holmes, Clive, Harris, Scott, Neal, James, Love, Seth, and Nicoll, James
- Published
- 2013
- Full Text
- View/download PDF
163. Consequences of beta-amyloid immunotherapy for Alzheimer's disease on neurons in the human cerebral cortex
- Author
-
Boche, Delphine, Amin, Jay, Mouton-Liger, François, Nasser, Mariam, Love, Seth, Gray, Francoise, Pickering, Ruth, Nicoll, James, Holmes, Clive, Hugon, Jacques, and Paquet, Claire
- Published
- 2013
- Full Text
- View/download PDF
164. Cerebral amyloid angiopathy-associated vasculopathy and apolipoprotein E accumulation after beta-amyloid immunotherapy for Alzheimer's disease
- Author
-
Nicoll, James, Sakai, Kenji, Love, Seth, and Boche, Delphine
- Published
- 2013
- Full Text
- View/download PDF
165. Virus load and neuropathology in the FIV model
- Author
-
Boche, Delphine, primary, Hurtrel, Maryse, additional, Gray, Francoise, additional, Claessens-maire, Marie-Annick, additional, Ganiere, Jean-Pierre, additional, Montagnier, Luc, additional, and Hurtrel, Bruno, additional
- Published
- 1996
- Full Text
- View/download PDF
166. Neuropathology of Early HIV‐1 Infection
- Author
-
Gray, Françoise, primary, Scaravilli, Francesco, additional, Everall, Ian, additional, Chretien, Fabrice, additional, An, Shu, additional, Boche, Delphine, additional, Adle‐Biassette, Homa, additional, Wingertsmann, Laure, additional, Durigon, Michel, additional, Hurtrel, Bruno, additional, Chiodi, Franceses, additional, Belli, Jeanne, additional, and Lantos, Peter, additional
- Published
- 1996
- Full Text
- View/download PDF
167. Effect of anti-cancer drugs on microglia in patient-derived breast cancer xenografted mouse models.
- Author
-
Paquet, Claire, Boche, Delphine, El Bouchtaoui, Morad, Gourmaud, Sarah, Janin, Anne, and Bousquet, Guilhem
- Subjects
- *
BREAST cancer , *DRUGS , *XENOGRAFTS - Abstract
A letter to the editor is presented in response to the article related to anti-cancer drugs effects on microglia and patient-derived xenograft models of breast cancer in the 2017 issue.
- Published
- 2017
- Full Text
- View/download PDF
168. Aβ Species Removal After Aβ42 Immunization.
- Author
-
Nicoll, James A. R., Barton, Edward, Boche, Delphine, Neal, Jim W., Ferrer, Isidro, Thompson, Petrina, Vlachouli, Christina, Wilkinson, David, Bayer, Antony, Games, Dora, Seubert, Peter, Schenk, Dale, and Holmes, Clive
- Published
- 2006
- Full Text
- View/download PDF
169. MotiQ: an open-source toolbox to quantify the cell motility and morphology of microglia
- Author
-
Hansen, Jan N., Brückner, Matthias, Pietrowski, Marie J., Jikeli, Jan F., Plescher, Monika, Beckert, Hannes, Schnaars, Mareike, Fülle, Lorenz, Reitmeier, Katharina, Langmann, Thomas, Förster, Irmgard, Boche, Delphine, Petzold, Gabor C., and Halle, Annett
- Abstract
MotiQ is an open-source software for automated quantification of microglial motility and morphology. MotiQ can be applied to in vivo, ex vivo, and in vitro data from confocal, epifluorescence, and two-photon microscopy. MotiQ is not limited to microglia—it can also be applied to other cell types.
- Published
- 2022
- Full Text
- View/download PDF
170. Lower microglial Fcγ RI/II and macrophage scavenger receptor a in human Alzheimer's disease after Aβ42 immunisation.
- Author
-
Zotova, Elina, Holmes, Clive, Neal, James, Love, Seth, Nicoll, James, and Boche, Delphine
- Published
- 2011
- Full Text
- View/download PDF
171. Tau pathology, neuroinflammation and adaptive immune cells in human primary tauopathies.
- Author
-
Hartnell, Iain James, Graffeuil, Manon, Mason, Luke, Marwaha, Pavan, Woodhouse, Declan, Jasper, William, Blum, David, Nicoll, James AR, Dorothee, Guillaume, and Boche, Delphine
- Abstract
Background: Primary tauopathies are neurodegenerative disorders that result from the accumulation of aggregated tau in the brain. Frontotemporal lobar degeneration with tau (FTLD‐tau) is a tauopathy that underlies ∼50% of cases of frontotemporal dementia (FTD). The discovery of genetic risk variants related to innate/adaptive immunity have highlighted a potential role for neuroinflammation in FTLD. Furthermore, studies have shown microglial and astrocyte activation together with T cell infiltration in the brain of THY‐Tau22 tauopathy mice. Methods: To study the relationship between tau and neuroinflammation we obtained FFPE brain tissue from 12 FTLD‐MAPT, 33 Pick's Disease (PiD), 45 Progressive Supranuclear Palsy (PSP) patients and 55 controls, via Brain UK. Using immunohistochemistry, we assessed tau pathology using antibodies 8 epitopes of tau with phosphorylation are different site. The immunophenotype of microglia were assessed with phenotypic markers and their morphology (ramified, reactive, ameboid) was analysed using Iba1. Astrocytic phenotype and T cell infiltration exploration is currently ongoing. Results: Tau epitopes AT8, AT100, PHF1, CP13 and Tau‐2 were increased in PiD, PSP and FTLD‐MAPT compared to controls, with expression levels significantly correlated between tau epitopes. Others are currently under analysis. Microglial markers Iba1, CD68, HLA‐DR and CD64 showed no difference between groups. However, CD16 was significantly increased in FTLD‐MAPT vs. control (p = 0.03) and correlations between tau and microglial markers (except with Iba1) were detected between disease groups. Morphological assessment revealed that ameboid microglia were the highest represented population in PiD and FTLD‐MAPT (p<0.0001 and p<0.0001, respectively); while reactive microglia were the most abundant population in PSP (p<0.0001). Preliminary observation of T cell staining confirms their recruitment in FTLD‐tau. Conclusion: These findings support the involvement of microglia in FTLD‐tau. Additional immuno‐phenotyping is necessary for further defining their role as well as T cell involvement in the disease pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
172. An integrated systems‐level analysis of the molecular changes resulting from systemic inflammation in Alzheimer's disease.
- Author
-
MacBean, Lachlan F, Smith, Rebecca G., Smith, Adam R., Nicoll, James AR, Boche, Delphine, and Lunnon, Katie
- Abstract
Background: In the current study we have performed an Epigenome‐Wide Association Study (EWAS) comparing bulk brain tissue in individuals with AD and non‐demented controls with or without a systemic infection, followed by targeted validation in isolated microglia. Method: DNA methylation is being quantified from 300 post‐mortem brain samples, 75 AD cases with infection, 75 AD cases without an infection, 75 non‐demented controls with infection and 75 non‐demented controls without an infection. DNA methylation of isolated microglia is also being quantified and gene expression data collected from the same samples to allow integration of epigenetic and transcriptomic data. Result: Statistical analyses performed using our established EWAS pipeline will identify significant differentially methylated positions. This is particularly expected to be observable in regions across genes implicated in disease pathology. Conclusion: Through cross‐examination of the four study groups we will identify novel patterns of methylation in those suffering from neuroinflammation not previously observed, also providing critical insight into the underlying molecular changes that arise in microglia as a result of neuroinflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
173. Aβ42 immunization reduces both tau and Aβ deposits in Alzheimer's disease
- Author
-
Boche, Delphine, Donald, Jane, Love, Seth, Neal, James W., Holmes, Clive, and Nicoll, James
- Published
- 2010
- Full Text
- View/download PDF
174. Processes underlying the vascular pathophysiology associated with Aβ immunization in Alzheimer's disease
- Author
-
Nicoll, James A.R., Hawkes, Cheryl, Holmes, Clive, Weller, Roy, Perry, Hugh, Boche, Delphine, Teeling, Jessica, and Carare, Roxana
- Published
- 2010
- Full Text
- View/download PDF
175. Aβ vaccination induces modifications of neuronal activated PKR staining in the brains of patients with Alzheimer's Disease (AD)
- Author
-
Paquet, Claire, Boche, Delphine, Gray, Françoise, Hugon, Jacques, and Nicoll, James
- Published
- 2009
- Full Text
- View/download PDF
176. The microglial translocator protein (TSPO) in Alzheimer's disease reflects a phagocytic phenotype.
- Author
-
Garland, Emma F., Antony, Henrike, Kulagowska, Laura, Scott, Thomas, Rogien, Charlotte, Bottlaender, Michel, Nicoll, James A. R., and Boche, Delphine
- Subjects
- *
ALZHEIMER'S disease , *TRANSLOCATOR proteins , *TEMPORAL lobe , *CEREBRAL cortex , *POSITRON emission tomography - Abstract
Translocator protein (TSPO) is a mitochondrial protein expressed by microglia, ligands for which are used as a marker of neuroinflammation in PET studies of Alzheimer's disease (AD). We previously showed increasing TSPO load in the cerebral cortex with AD progression, consistent with TSPO PET scan findings. Here, we aim to characterise the microglial phenotype associated with TSPO expression to aid interpretation of the signal generated by TSPO ligands in patients. Human post-mortem sections of temporal lobe (TL) and cerebellum (Cb) from cases classified by Braak group (0–II, III–IV, V–VI; each n = 10) were fluorescently double labelled for TSPO and microglial markers: Iba1, HLA-DR, CD68, MSR-A and CD64. Quantification was performed on scanned images using QuPath software to assess the microglial phenotype of TSPO. Qualitative analysis was also performed for TSPO with GFAP (astrocytes), CD31 (endothelial cells) and CD163 (perivascular macrophages) to characterise the cellular profile of TSPO. The percentage of CD68+TSPO+ double-labelled cells was significantly higher than for other microglial markers in both brain regions and in all Braak stages, followed by MSR-A+TSPO+ microglia. Iba1+TSPO+ cells were more numerous in the cerebellum than the temporal lobe, while CD64+TSPO+ cells were more numerous in the temporal lobe. No differences were observed for the other microglial markers. TSPO expression was also detected in endothelial cells, but not detected in astrocytes nor in perivascular macrophages. Our data suggest that TSPO is mainly related to a phagocytic profile of microglia (CD68+) in human AD, potentially highlighting the ongoing neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
177. No replicating evidence for anti‐amyloid‐β autoantibodies in cerebral amyloid angiopathy‐related inflammation.
- Author
-
van den Berg, Emma, Roelofs, Rian, Jäkel, Lieke, Greenberg, Steven M., Charidimou, Andreas, van Etten, Ellis S., Boche, Delphine, Klijn, Catharina J. M., Schreuder, Floris H. B. M., Kuiperij, H. Bea, and Verbeek, Marcel M.
- Subjects
- *
PLASMA cells , *CEREBROSPINAL fluid , *IMMUNOGLOBULIN G , *AUTOANTIBODIES , *CEREBRAL amyloid angiopathy - Abstract
Objective: Elevated levels of anti‐amyloid‐β (anti‐Aβ) autoantibodies in cerebrospinal fluid (CSF) have been proposed as a diagnostic biomarker for cerebral amyloid angiopathy‐related inflammation (CAA‐RI). We aimed to independently validate the immunoassay for quantifying these antibodies and evaluate its diagnostic value for CAA‐RI. Methods: We replicated the immunoassay to detect CSF anti‐Aβ autoantibodies using CSF from CAA‐RI patients and non‐CAA controls with unrelated disorders and further characterized its performance. Moreover, we conducted a literature review of CAA‐RI case reports to investigate neuropathological and CSF evidence of the nature of the inflammatory reaction in CAA‐RI. Results: The assay demonstrated a high background signal in CSF, which increased and corresponded with higher total immunoglobulin G (IgG) concentration in CSF (rsp = 0.51, p = 0.02). Assay levels were not elevated in CAA‐RI patients (n = 6) compared to non‐CAA controls (n = 20; p = 0.64). Literature review indicated only occasional presence of B‐lymphocytes and plasma cells (i.e., antibody‐producing cells), alongside the abundant presence of activated microglial cells, T‐cells, and other monocyte lineage cells. CSF analysis did not convincingly indicate intrathecal IgG production. Interpretation: We were unable to reproduce the reported elevation of anti‐Aβ autoantibody concentration in CSF of CAA‐RI patients. Our findings instead support nonspecific detection of IgG levels in CSF by the assay. Reviewed CAA‐RI case reports suggested a widespread cerebral inflammatory reaction. In conclusion, our findings do not support anti‐Aβ autoantibodies as a diagnostic biomarker for CAA‐RI. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
178. Mini-symposium: Role of the inflammasome in brain pathogenesis: a potential therapeutic target? - introduction.
- Author
-
Boche, Delphine and Brough, David
- Subjects
- *
INFLAMMASOMES , *MOLECULAR structure , *STROKE - Abstract
An introduction is presented in which the editor discusses various reports within the issue on topics including inflammasome complex as an initial host defense against infections, molecular structure of inflammasome, and stroke.
- Published
- 2017
- Full Text
- View/download PDF
179. Microglial motility in Alzheimer's disease and after Aβ42 immunotherapy: a human post-mortem study.
- Author
-
Franco-Bocanegra, Diana K., George, Bethany, Lau, Laurie C., Holmes, Clive, Nicoll, James A. R., and Boche, Delphine
- Subjects
ALZHEIMER'S disease ,AUTOPSY ,HUMAN experimentation ,CELL motility ,IMMUNOTHERAPY ,PROTEIN expression - Abstract
Microglial function is highly dependent on cell motility, with baseline motility required for homeostatic surveillance activity and directed motility to migrate towards a source of injury. Experimental evidence suggests impaired microglial motility in Alzheimer's disease (AD) and therefore we have investigated whether the expression of proteins associated with motility is altered in AD and affected by the Aβ immunotherapy using post-mortem brain tissue of 32 controls, 44 AD cases, and 16 AD cases from our unique group of patients immunised against Aβ42 (iAD). Sections of brain were immunolabelled and quantified for (i) the motility-related microglial proteins Iba1, cofilin 1 (CFL1), coronin-1a (CORO1A) and P2RY12, and (ii) pan-Aβ, Aβ42 and phosphorylated tau (ptau). The neuroinflammatory environment was characterised using Meso Scale Discovery multiplex assays. The expression of all four motility-related proteins was unmodified in AD compared with controls, whereas Iba1 and P2RY12, the homeostatic markers, were increased in the iAD group compared with AD. Iba1 and P2RY12 showed significant positive correlations with Aβ in controls but not in the AD or iAD groups. Pro- and anti-inflammatory proteins were increased in AD, whereas immunotherapy appears to result in a slightly less pro-inflammatory environment. Our findings suggest that as Aβ appears during the ageing process, the homeostatic Iba1 and P2RY12 –positive microglia respond to Aβ, but this response is absent in AD. Aβ-immunisation promoted increased Iba1 and P2RY12 expression, likely reflecting increased baseline microglial motility but without restoring the profile observed in controls. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
180. Aβ43 in human Alzheimer's disease: effects of active Aβ42 immunization.
- Author
-
Jäkel, Lieke, Boche, Delphine, Nicoll, James A. R., and Verbeek, Marcel M.
- Subjects
- *
ALZHEIMER'S disease , *CEREBRAL amyloid angiopathy , *IMMUNIZATION , *ALZHEIMER'S patients , *AMYLOID beta-protein - Abstract
Neuropathological follow-up of patients with Alzheimer's disease (AD) who participated in the first clinical trial of Amyloid-β 42 (Aβ42) immunization (AN1792, Elan Pharmaceuticals) has shown that immunization can induce removal of Aβ42 and Aβ40 from plaques, whereas analysis of the cerebral vessels has shown increased levels of these Aβ peptides in cerebral amyloid angiopathy (CAA). Aβ43 has been less frequently studied in AD, but its aggregation propensity and neurotoxic properties suggest it may have an important pathogenic role. In the current study we show by using immunohistochemistry that in unimmunized AD patients Aβ43 is a frequent constituent of plaques (6.0% immunostained area), similar to Aβ42 (3.9% immunostained area). Aβ43 immunostained area was significantly higher than that of Aβ40 (2.3%, p = 0.006). In addition, we show that Aβ43 is only a minor component of CAA in both parenchymal vessels (1.5 Aβ43-positive vessels per cm2 cortex vs. 5.3 Aβ42-positive vessels, p = 0.03, and 6.2 Aβ40-positive vessels, p = 0.045) and leptomeningeal vessels (5.6% Aβ43-positive vessels vs. 17.3% Aβ42-positive vessels, p = 0.007, and 27.4% Aβ40-positive vessels, p = 0.003). Furthermore, we have shown that Aβ43 is cleared from plaques after Aβ immunotherapy, similar to Aβ42 and Aβ40. Cerebrovascular Aβ43 levels did not change after immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
181. Molecular Mechanisms of Microglial Motility: Changes in Ageing and Alzheimer's Disease.
- Author
-
Franco-Bocanegra, Diana K., McAuley, Ciaran, Nicoll, James A. R., and Boche, Delphine
- Subjects
MICROGLIA ,ALZHEIMER'S disease ,PURINERGIC receptors ,MEMBRANE proteins ,CENTRAL nervous system ,CYTOSKELETON - Abstract
Microglia are the tissue-resident immune cells of the central nervous system, where they constitute the first line of defense against any pathogens or injury. Microglia are highly motile cells and in order to carry out their function, they constantly undergo changes in their morphology to adapt to their environment. The microglial motility and morphological versatility are the result of a complex molecular machinery, mainly composed of mechanisms of organization of the actin cytoskeleton, coupled with a "sensory" system of membrane receptors that allow the cells to perceive changes in their microenvironment and modulate their responses. Evidence points to microglia as accountable for some of the changes observed in the brain during ageing, and microglia have a role in the development of neurodegenerative diseases, such as Alzheimer's disease. The present review describes in detail the main mechanisms driving microglial motility in physiological conditions, namely, the cytoskeletal actin dynamics, with emphasis in proteins highly expressed in microglia, and the role of chemotactic membrane proteins, such as the fractalkine and purinergic receptors. The review further delves into the changes occurring to the involved proteins and pathways specifically during ageing and in Alzheimer's disease, analyzing how these changes might participate in the development of this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
182. Additional file 1: of Pyroglutamate and Isoaspartate modified Amyloid-Beta in ageing and Alzheimer’s disease
- Author
-
Moro, Maria, Phillips, Andrew, Gaimster, Katie, Paul, Christian, Amritpal Mudher, Nicoll, James, and Boche, Delphine
- Subjects
3. Good health - Abstract
Contains all supplemental information mentioned in this manuscript. Table S1. Characteristics of the old control and AD cohorts. Figure S1. Representation of the correlations between pE3-Aβ and phospho-TAU in OC and AD groups together. (DOC 203 kb)
183. Additional file 1: of Pyroglutamate and Isoaspartate modified Amyloid-Beta in ageing and Alzheimer’s disease
- Author
-
Moro, Maria, Phillips, Andrew, Gaimster, Katie, Paul, Christian, Amritpal Mudher, Nicoll, James, and Boche, Delphine
- Subjects
3. Good health - Abstract
Contains all supplemental information mentioned in this manuscript. Table S1. Characteristics of the old control and AD cohorts. Figure S1. Representation of the correlations between pE3-Aβ and phospho-TAU in OC and AD groups together. (DOC 203 kb)
184. Systemic infection modifies the neuroinflammatory response in late stage Alzheimer’s disease.
- Author
-
Rakic, Sonja, Hung, Yat M. A., Smith, Matthew, So, Denise, Tayler, Hannah M., Varney, William, Wild, Joe, Harris, Scott, Holmes, Clive, Love, Seth, Stewart, William, Nicoll, James A. R., and Boche, Delphine
- Subjects
INFLAMMATION ,ALZHEIMER'S patients ,ANIMAL models in research - Abstract
Clinical studies indicate that systemic infections accelerate cognitive decline in Alzheimer’s disease. Animal models suggest that this may be due to enhanced pro-inflammatory changes in the brain. We have performed a post-mortem human study to determine whether systemic infection modifies the neuropathology and in particular, neuroinflammation, in the late-stage of the disease. Sections of cerebral cortex and underlying white matter from controls and Alzheimer's patients who died with or without a terminal systemic infection were immunolabelled and quantified for: (i) Αβ and phosphorylated-tau; (ii) the inflammation-related proteins Iba1, CD68, HLA-DR, FcγRs (CD64, CD32a, CD32b, CD16), CHIL3L1, IL4R and CCR2; and (iii) T-cell marker CD3. In Alzheimer's disease, the synaptic proteins synaptophysin and PSD-95 were quantified by ELISA, and the inflammatory proteins and mRNAs by MesoScale Discovery Multiplex Assays and qPCR, respectively. Systemic infection in Alzheimer's disease was associated with decreased CD16 (p = 0.027, grey matter) and CD68 (p = 0.015, white matter); increased CD64 (p = 0.017, white matter) as well as increased protein expression of IL6 (p = 0.047) and decreased IL5 (p = 0.007), IL7 (p = 0.002), IL12/IL23p40 (p = 0.001), IL15 (p = 0.008), IL16 (p < 0.001) and IL17A (p < 0.001). Increased expression of anti-inflammatory genes CHI3L1 (p = 0.012) and IL4R (p = 0.004) were detected in this group. T-cell recruitment to the brain was reduced when systemic infection was present. However, exposure to systemic infection did not modify the pathology. In Alzheimer's disease, CD68 (p = 0.026), CD64 (p = 0.002), CHI3L1 (p = 0.016), IL4R (p = 0.005) and CCR2 (p = 0.010) were increased independently of systemic infection. Our findings suggest that systemic infections modify neuroinflammatory processes in Alzheimer's disease. However, rather than promoting pro-inflammatory changes, as observed in experimental models, they seem to promote an anti-inflammatory, potentially immunosuppressive, environment in the human brain. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
185. Glial reactivity and T cell infiltration in frontotemporal lobar degeneration with tau pathology.
- Author
-
Hartnell, Iain J, Woodhouse, Declan, Jasper, William, Mason, Luke, Marwaha, Pavan, Graffeuil, Manon, Lau, Laurie C, Norman, Jeanette L, Chatelet, David S, Buee, Luc, Nicoll, James A R, Blum, David, Dorothee, Guillaume, and Boche, Delphine
- Subjects
- *
PROGRESSIVE supranuclear palsy , *FRONTOTEMPORAL lobar degeneration , *DEGENERATION (Pathology) , *T cells , *FRONTOTEMPORAL dementia , *GLUTAMINE synthetase - Abstract
Frontotemporal lobar degeneration with tau (FTLD-tau) is a group of tauopathies that underlie ∼50% of FTLD cases. Identification of genetic risk variants related to innate/adaptive immunity have highlighted a role for neuroinflammation and neuroimmune interactions in FTLD. Studies have shown microglial and astrocyte activation together with T cell infiltration in the brain of THY-Tau22 tauopathy mice. However, this remains to be confirmed in FTLD-tau patients. We conducted a detailed post-mortem study of FTLD-tau cases including 45 progressive supranuclear palsy with clinical frontotemporal dementia, 33 Pick's disease, 12 FTLD-MAPT and 52 control brains to characterize the link between phosphorylated tau (pTau) epitopes and the innate and adaptive immunity. Tau pathology was assessed in the cerebral cortex using antibodies directed against: Tau-2 (phosphorylated and unphosphorylated tau), AT8 (pSer202/pThr205), AT100 (pThr212/pSer214), CP13 (pSer202), PHF1 (pSer396/pSer404), pThr181 and pSer356. The immunophenotypes of microglia and astrocytes were assessed with phenotypic markers (Iba1, CD68, HLA-DR, CD64, CD32a, CD16 for microglia and GFAP, EAAT2, glutamine synthetase and ALDH1L1 for astrocytes). The adaptive immune response was explored via CD4+ and CD8+ T cell quantification and the neuroinflammatory environment was investigated via the expression of 30 inflammatory-related proteins using V-Plex Meso Scale Discovery. As expected, all pTau markers were increased in FTLD-tau cases compared to controls. pSer356 expression was greatest in FTLD-MAPT cases versus controls (P < 0.0001), whereas the expression of other markers was highest in Pick's disease. Progressive supranuclear palsy with frontotemporal dementia consistently had a lower pTau protein load compared to Pick's disease across tau epitopes. The only microglial marker increased in FTLD-tau was CD16 (P = 0.0292) and specifically in FTLD-MAPT cases (P = 0.0150). However, several associations were detected between pTau epitopes and microglia, supporting an interplay between them. GFAP expression was increased in FTLD-tau (P = 0.0345) with the highest expression in Pick's disease (P = 0.0019), while ALDH1L1 was unchanged. Markers of astrocyte glutamate cycling function were reduced in FTLD-tau (P = 0.0075; Pick's disease: P < 0.0400) implying astrocyte reactivity associated with a decreased glutamate cycling activity, which was further associated with pTau expression. Of the inflammatory proteins assessed in the brain, five chemokines were upregulated in Pick's disease cases (P < 0.0400), consistent with the recruitment of CD4+ (P = 0.0109) and CD8+ (P = 0.0014) T cells. Of note, the CD8+ T cell infiltration was associated with pTau epitopes and microglial and astrocytic markers. Our results highlight that FTLD-tau is associated with astrocyte reactivity, remarkably little activation of microglia, but involvement of adaptive immunity in the form of chemokine-driven recruitment of T lymphocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
186. Changes in the locus coeruleus during the course of Alzheimer's disease and their relationship to cortical pathology.
- Author
-
Beardmore, Rebecca, Durkin, Matthew, Zayee‐Mellick, Faizan, Lau, Laurie C., Nicoll, James A. R., Holmes, Clive, and Boche, Delphine
- Subjects
- *
LOCUS coeruleus , *ALZHEIMER'S disease , *TYROSINE hydroxylase , *PATHOLOGY , *DISEASE progression , *TEMPORAL lobe - Abstract
Aims: In Alzheimer's disease (AD), the locus coeruleus (LC) undergoes early and extensive neuronal loss, preceded by abnormal intracellular tau aggregation, decades before the onset of clinical disease. Neuromelanin‐sensitive MRI has been proposed as a method to image these changes during life. Surprisingly, human post‐mortem studies have not examined how changes in LC during the course of the disease relate to cerebral pathology following the loss of the LC projection to the cortex. Methods: Immunohistochemistry was used to examine markers for 4G8 (pan‐Aβ) and AT8 (ptau), LC integrity (neuromelanin, dopamine β‐hydroxylase [DβH], tyrosine hydroxylase [TH]) and microglia (Iba1, CD68, HLA‐DR) in the LC and related temporal lobe pathology of 59 post‐mortem brains grouped by disease severity determined by Braak stage (0‐II, III‐IV and V‐VI). The inflammatory environment was assessed using multiplex assays. Results: Changes in the LC with increasing Braak stage included increased neuronal loss (p < 0.001) and microglial Iba1 (p = 0.005) together with a reduction in neuromelanin (p < 0.001), DβH (p = 0.002) and TH (p = 0.041). Interestingly in LC, increased ptau and loss of neuromelanin were detected from Braak stage III‐IV (p = 0.001). At Braak stage V/VI, the inflammatory environment was different in the LC vs TL, highlighting the anatomical heterogeneity of the inflammatory response. Conclusions: Here, we report the first quantification of neuromelanin during the course of AD and its relationship to AD pathology and neuroinflammation in the TL. Our findings of neuromelanin loss early in AD and before the neuroinflammatory reaction support the use of neuromelanin‐MRI as a sensitive technique to identify early changes in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
187. Innate immunity in Alzheimer’s disease: the relevance of animal models?
- Author
-
Franco Bocanegra, Diana K., Nicoll, James A. R., and Boche, Delphine
- Subjects
- *
ALZHEIMER'S disease treatment , *NATURAL immunity , *CELL morphology , *CELL metabolism , *ETIOLOGY of diseases , *ANIMAL models in research - Abstract
The mouse is one of the organisms most widely used as an animal model in biomedical research, due to the particular ease with which it can be handled and reproduced in laboratory. As a member of the mammalian class, mice share with humans many features regarding metabolic pathways, cell morphology and anatomy. However, important biological differences between mice and humans exist and must be taken into consideration when interpreting research results, to properly translate evidence from experimental studies into information that can be useful for human disease prevention and/or treatment. With respect to Alzheimer’s disease (AD), much of the experimental information currently known about this disease has been gathered from studies using mainly mice as models. Therefore, it is notably important to fully characterise the differences between mice and humans regarding important aspects of the disease. It is now widely known that inflammation plays an important role in the development of AD, a role that is not only a response to the surrounding pathological environment, but rather seems to be strongly implicated in the aetiology of the disease as indicated by the genetic studies. This review highlights relevant differences in inflammation and in microglia, the innate immune cell of the brain, between mice and humans regarding genetics and morphology in normal ageing, and the relationship of microglia with AD-like pathology, the inflammatory profile, and cognition. We conclude that some noteworthy differences exist between mice and humans regarding microglial characteristics, in distribution, gene expression, and states of activation. This may have repercussions in the way that transgenic mice respond to, and influence, the AD-like pathology. However, despite these differences, human and mouse microglia also show similarities in morphology and behaviour, such that the mouse is a suitable model for studying the role of microglia, as long as these differences are taken into consideration when delineating new strategies to approach the study of neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
188. The mitochondrial protein TSPO in Alzheimer's disease: relation to the severity of AD pathology and the neuroinflammatory environment.
- Author
-
Garland, Emma F., Dennett, Oliver, Lau, Laurie C., Chatelet, David S., Bottlaender, Michel, Nicoll, James A. R., and Boche, Delphine
- Subjects
- *
ALZHEIMER'S disease , *MITOCHONDRIAL pathology , *MITOCHONDRIAL proteins , *CEREBELLAR cortex , *TEMPORAL lobe , *CEREBRAL cortex - Abstract
The 18kD translocator protein (TSPO) is used as a positron emission tomography (PET) target to quantify neuroinflammation in patients. In Alzheimer's disease (AD), the cerebellum is the pseudo-reference region for comparison with the cerebral cortex due to the absence of AD pathology and lower levels of TSPO. However, using the cerebellum as a pseudo-reference region is debated, with other brain regions suggested as more suitable. This paper aimed to establish the neuroinflammatory differences between the temporal cortex and cerebellar cortex, including TSPO expression. Using 60 human post-mortem samples encompassing the spectrum of Braak stages (I–VI), immunostaining for pan-Aβ, hyperphosphorylated (p)Tau, TSPO and microglial proteins Iba1, HLA–DR and MSR-A was performed in the temporal cortex and cerebellum. In the cerebellum, Aβ but not pTau, increased over the course of the disease, in contrast to the temporal cortex, where both proteins were significantly increased. TSPO increased in the temporal cortex, more than twofold in the later stages of AD compared to the early stages, but not in the cerebellum. Conversely, Iba1 increased in the cerebellum, but not in the temporal cortex. TSPO was associated with pTau in the temporal cortex, suggesting that TSPO positive microglia may be reacting to pTau itself and/or neurodegeneration at later stages of AD. Furthermore, the neuroinflammatory microenvironment was examined, using MesoScale Discovery assays, and IL15 only was significantly increased in the temporal cortex. Together this data suggests that the cerebellum maintains a more homeostatic environment compared to the temporal cortex, with a consistent TSPO expression, supporting its use as a pseudo-reference region for quantification in TSPO PET scans. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
189. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies.
- Author
-
De Picker, Livia J., Morrens, Manuel, Branchi, Igor, Haarman, Bartholomeus C.M., Terada, Tatsuhiro, Kang, Min Su, Boche, Delphine, Tremblay, Marie-Eve, Leroy, Claire, Bottlaender, Michel, and Ottoy, Julie
- Subjects
- *
ENCEPHALITIS , *BRAIN imaging , *ALZHEIMER'S disease , *CASE-control method , *POSITRON emission tomography - Abstract
• Widespread TSPO PET signal increases in cortical grey matter in AD and other neurodegenerative disorder. • Cortico-limbic signal increases in AD, MCI , other neurodegenerative disorders, mood disorders, and MS. • Thalamic signal increases in AD, other neurodegenerative disorders, MS, and chronic pain and functional disorders. • Quantification method (V T vs. reference tissue models) accounted for 25% of transdiagnostic between-study variability. • Patient age and radioligand generation accounted for 9 and 5% of transdiagnostic between-study variability. The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, P FDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, P FDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, P FDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all P FDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; V T -based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to V T -based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
190. T Lymphocytes and Their Potential Role in Dementia with Lewy Bodies.
- Author
-
Amin, Jay, Gee, Claire, Stowell, Kiran, Coulthard, Daisy, and Boche, Delphine
- Subjects
- *
LEWY body dementia , *ALZHEIMER'S disease , *T cells , *CEREBROSPINAL fluid , *PARKINSON'S disease , *POSTMORTEM changes - Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative cause of dementia. People with DLB have an inferior prognosis compared to Alzheimer's disease (AD), but the diseases overlap in their neuropathology and clinical syndrome. It is imperative that we enhance our understanding of the aetiology and pathogenesis of DLB. The impact of peripheral inflammation on the brain in dementia has been increasingly explored in recent years, with T lymphocyte recruitment into brain parenchyma identified in AD and Parkinson's disease. There is now a growing range of literature emerging on the potential role of innate and adaptive immune cells in DLB, including T lymphocytes. In this review, we examine the profile of T lymphocytes in DLB, focusing on studies of post-mortem brain tissue, cerebrospinal fluid, and the blood compartment. We present an integrated viewpoint on the results of these studies by proposing how changes to the T lymphocyte profile in the brain and periphery may relate to each other. Improving our understanding of T lymphocytes in DLB has the potential to guide the development of disease-modifying treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
191. Microglial motility and morphology in Alzheimer's disease, and after Aβ42-immunotherapy
- Author
-
Franco Bocanegra, Diana Karina, Boche, Delphine, and Nicoll, James
- Subjects
616.8 - Abstract
Microglia are the resident immune cells of the brain. Their main functions in the adult brain are to provide protection against pathogens and to remove cellular debris via phagocytosis. Microglial function is highly dependent on cell motility. Evidence from Alzheimer's disease (AD) animal models supports microglial motility impairment in AD, possibly linked to Aβ accumulation. In AD patients immunised against Aβ42, Aβ plaque removal is partly due to phagocytic microglia, supporting an association between microglial function and Aβ clearance. The aim of the present study is to evaluate the expression of microglial motility-related proteins, as well as microglial morphological features, in the inferior parietal lobule of 32 controls, 44 AD cases and 16 immunised AD (iAD) cases, patients who had been immunised against the Aβ42 peptide. Immunohistochemistry (IHC) was performed for the proteins Iba1, CFL1, CORO1A and P2RY12, and protein load was assessed with an automated image analysis method. The neuroinflammatory environment was evaluated with Meso Scale Discovery protein immunoassays. Iba1 IHC was also used for microglial cell counting and morphological analysis. Fluorescent Iba1 IHC, confocal microscopy and semi-automated 3Dreconstruction were used for detailed assessment of microglial morphological features. Among the most relevant results, the levels of the motility-related proteins studied showed no difference in AD cases compared to controls. In the iAD group compared to AD, the levels of Iba1 were increased in grey and white matter, and the level of P2RY12 in the grey matter only. There was no difference in the total number of microglia in AD compared to controls, but there was an increase in iAD compared to AD. The ramified population was reduced in AD compared to controls, while in iAD both the ramified and amoeboid populations were increased. The microglial cell body volume was increased in AD compared to controls, whereas the total process length was increased in iAD compared to AD. Our results highlight some potentially relevant effects of the disease, and of Aβ immunotherapy on microglial motility, and help further our understanding of the functional spectrum of microglia in health and disease.
- Published
- 2020
192. Inflammation in Dementia with Lewy bodies and Alzheimer's disease
- Author
-
Amin, Jay, Boche, Delphine, and Holmes, Clive
- Subjects
616.8 - Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative cause of dementia. However, the aetiology of DLB remains poorly understood in comparison with Alzheimer's disease (AD) and Parkinson's disease (PD). Current evidence supports that neuroinflammation, with involvement of the peripheral immune system, occurs in both AD and PD. Genetic studies in particular support an aetiological role for inflammation in AD rather than it being merely a consequence of neurodegeneration. Despite extensive research into the role of inflammation in AD and PD, there have been a paucity of studies in DLB. I hypothesised that DLB would show a specific cerebral and systemic inflammatory profile. In order to investigate this hypothesis, two studies were performed. A cross-sectional clinical study investigated peripheral inflammation in DLB, AD and controls using flow cytometry and multiplex immunoassay, and post-mortem human brain tissue work examined microglial immunophenotype in DLB, AD and controls using immunohistochemistry. The clinical study revealed increased serum concentrations of two proinflammatory cytokines (IL1β and IL6) in DLB compared with controls. In addition, flow cytometry showed a decline in cell populations associated with adaptive immunity (helper T cells and activated B cells) in DLB compared to AD. These data demonstrate senescence of the adaptive immune system in DLB compared with AD, possibly driving a chronic inflammatory state. The post-mortem work confirmed increased cerebral protein deposition in DLB and AD, but the two diseases showed markedly different microglial phenotypes. AD was characterised by a strong phagocytic microglial phenotype, but in DLB there was no evidence of increased activation of any phenotype. These findings may be associated with the different profiles of the peripheral adaptive immune system, with AD characterised by increased antibody-mediated microglial activation compared with DLB. The two studies undertaken as part of this project appear to show that the immunophenotype of DLB is distinct from that of AD, with cerebral inflammation not a primary feature of DLB as it is in AD. This has therapeutic implications in that the use of anti-inflammatory therapy may not indicated in DLB. Furthermore, identification of a unique peripheral immune profile in DLB warrants further exploration in order to develop a blood-based immune biomarker that could differentiate these two diseases.
- Published
- 2019
193. Haptoglobin as a novel treatment for subarachnoid haemorrhage
- Author
-
Morton, Matthew, Galea, Ian, and Boche, Delphine
- Subjects
616.8 - Abstract
Subarachnoid haemorrhage (SAH) is a severe neurological condition caused by rupture of an intracranial aneurysm. A clot forms in the subarachnoid space and erythrocyte lysis causes haemoglobin (Hb) to accumulate in the CSF. In patients surviving the initial bleed, secondary complications such as vasospasm and delayed ischaemic neurological deficits (DIND) can result in poor long term outcome. Hb is neurotoxic through oxidative mechanisms and has also been implicated in the pathogenesis of vasospasm. Management of SAH patients is aimed at securing the aneurysm, inhibiting vasospasm and diversion of CSF to treat hydrocephalus; high morbidity means novel treatments to improve long term outcome are needed and there are none targeting Hb toxicity. Haptoglobin (Hp) is responsible for binding to cell free Hb, facilitating scavenging by CD163-positive macrophages. Hp inhibits free Hb’s pro-oxidative potential by shielding the redox active iron contained in the haem group, rendering Hb oxidatively inert. Two co-dominant HP alleles (HP1 and HP2) exist in humans as a result of an intragenic duplication event. This lead to the production of dimers in HP1 homozygotes (HP1-1), dimers and trimers in heterozygotes (HP2-1), and finally tetramers and large polymers in HP2 homozygotes (HP2-2). Hp isoforms differ in their expression levels, their ability to inhibit Hb’s oxidative potential, and when in complex with Hb, their affinity for CD163 and uptake by macrophages. In this study I hypothesized that: haptoglobin affects outcome after subarachnoid haemorrhage through its role in haemoglobin scavenging and haptoglobin supplementation after SAH is of therapeutic benefit. The aims were to: 1) investigate if HP genotype influences outcome after SAH via its role in Hb scavenging, 2) characterise the kinetics of Hb scavenging in the two week period after SAH, 3) establish whether Hp can reverse Hb neurotoxicity in vitro, and 4) establish and characterise an in vivo model of SAH to translate the results of the in vitro experiments. Due to differential Hb scavenging function, it was hypothesized that HP genotype impacts on outcome after SAH. 1299 SAH patients were typed for their HP duplication genotype and the single nucleotide polymorphism rs2000999, the largest genetic contributor to Hp expression level. Logistic regression analysis of long term outcome identified no effect of rs2000999 but the Hp duplication affected outcome, implying mediation through Hp function rather than expression level. HP2-2 was associated with favourable outcome in high Fisher grade patients (grades III and IV, Odds Ratio (OR): 2.4, 95% Confidence Intervals (CI) 1.4-.4.3, p=0.007). To seek further mechanistic evidence for the effect of Hp on outcome after SAH, the kinetics of Hb and Hp was investigated in serial cerebrospinal fluid (CSF) samples from 44 Fisher grade III-IV SAH patients during a two week period post-ictus. Total Hb levels increased gradually, reaching a plateau of 11.5μM (median between days 11 and 13, interquartile range (IQR): 2.1-16.2μM); the majority of this Hb was found to be uncomplexed (median 96.3%, (IQR): 83.3-99.4%, from third day onwards). There was a trend towards an association of high uncomplexed Hb levels with worse outcome (modified Rankin Scale (mRS) at 6 months post-ictus) (OR: 0.7, 95% CI 0.5-1.0, p=0.064). Auto-oxidation of Hb can cause oxidative damage which inhibits complex formation by Hp. Exogenous Hp was added to the CSF, enough to saturate all Hb, to investigate how much Hb was still able to bind Hp; 90.6% (IQR: 65.8- 96.5%) of the uncomplexed Hb was able to form complexes with exogenous Hp. The Hb scavenging ability of Hp isoforms was investigated with linear mixed modelling. Significantly lower levels of uncomplexed Hb were associated with the Hp2-2 phenotype (p=0.03, vs Hp1-1), in keeping with improved Hb scavenging due to Hp2-2’s higher valency for Hb. To test if Hp can protect against Hb neurotoxicity, primary hippocampal neurones from C57BL/6 mouse pups were cultured and treated with Hb, Hb+Hp, Hp or vehicle. Following a week in culture, viable neurones were counted. Hb produced a dose dependent cytotoxic effect (pSignificantly lower levels of uncomplexed Hb were associated with the Hp2-2 phenotype (p=0.03, vs Hp1-1), in keeping with improved Hb scavenging due to Hp2-2’s higher valency for Hb. To test if Hp can protect against Hb neurotoxicity, primary hippocampal neurones from C57BL/6 mouse pups were cultured and treated with Hb, Hb+Hp, Hp or vehicle. Following a week in culture, viable neurones were counted. Hb produced a dose dependent cytotoxic effect (p=0.003). No adverse effects on neuronal viability were observed with Hp alone. To translate the in vitro findings, I established in our laboratory an in vivo murine model of SAH, involving a stereotaxic injection of blood into the pre-chiasmatic cistern of C57BL/6 mice. The model was optimized to refine the technique, to prepare for future experiments testing Hp as a therapeutic agent after experimental SAH.
- Published
- 2018
194. Differential perivascular microglial activation in the deep white matter in vascular dementia developed post‐stroke.
- Author
-
Hase, Yoshiki, Ameen‐Ali, Kamar E., Waller, Rachel, Simpson, Julie E., Stafford, Charlotte, Mahesh, Ayushi, Ryan, Lucy, Pickering, Lucy, Bodman, Caroline, Hase, Mai, Boche, Delphine, Horsburgh, Karen, Wharton, Stephen B., and Kalaria, Raj N.
- Subjects
- *
WHITE matter (Nerve tissue) , *VASCULAR dementia , *MICROGLIA , *MYELIN basic protein , *FRONTAL lobe , *MYELOID cells - Abstract
With the hypothesis that perivascular microglia are involved as neuroinflammatory components of the gliovascular unit contributing to white matter hyperintensities on MRI and pathophysiology, we assessed their status in stroke survivors who develop dementia. Immunohistochemical and immunofluorescent methods were used to assess the distribution and quantification of total and perivascular microglial cell densities in 68 brains focusing on the frontal lobe WM and overlying neocortex in post‐stroke dementia (PSD), post‐stroke non‐dementia (PSND) and similar age control subjects. We primarily used CD68 as a marker of phagocytic microglia, as well as other markers of microglia including Iba‐1 and TMEM119, and the myeloid cell marker TREM2 to assess dementia‐specific changes. We first noted greater total densities of CD68+ and TREM2+ cells per mm2 in the frontal WM compared to the overlying cortex across the stroke cases and controls (p = 0.001). PSD subjects showed increased percentage of activated perivascular CD68+ cells distinct from ramified or primed microglia in the WM (p < 0.05). However, there was no apparent change in perivascular TREM2+ cells. Total densities of TREM2+ cells were only ~10% of CD68+ cells but there was high degree of overlap (>70%) between them in both the WM and the cortex. CD68 and Iba‐1 or CD68 and TMEM119 markers were colocalised by ~55%. Within the deep WM, ~30% of CD68+ cells were co‐localised with fragments of degraded myelin basic protein. Among fragmented CD68+ cells in adjacent WM of PSD subjects, >80% of the cells expressed cleaved caspase‐3. Our observations suggest although the overall repertoire of perivascular microglial cells is not changed in the parenchyma, PSD subjects accrue more perivascular‐activated CD68+ microglia rather than TREM2+ cells. This implies there is a subset of CD68+ cells, which are responsible for the differential response in perivascular inflammation within the gliovascular unit of the deep WM. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
195. Combination Therapy in Alzheimer's Disease: Is It Time?
- Author
-
Salehipour, Arash, Bagheri, Motahareh, Sabahi, Mohammadmahdi, Dolatshahi, Mahsa, and Boche, Delphine
- Subjects
- *
ALZHEIMER'S disease , *TREATMENT effectiveness , *COMBINED modality therapy , *PEPTIDES - Abstract
Alzheimer's disease (AD) is the most common cause of dementia globally. There is increasing evidence showing AD has no single pathogenic mechanism, and thus treatment approaches focusing only on one mechanism are unlikely to be meaningfully effective. With only one potentially disease modifying treatment approved, targeting amyloid-β (Aβ), AD is underserved regarding effective drug treatments. Combining multiple drugs or designing treatments that target multiple pathways could be an effective therapeutic approach. Considering the distinction between added and combination therapies, one can conclude that most trials fall under the category of added therapies. For combination therapy to have an actual impact on the course of AD, it is likely necessary to target multiple mechanisms including but not limited to Aβ and tau pathology. Several challenges have to be addressed regarding combination therapy, including choosing the correct agents, the best time and stage of AD to intervene, designing and providing proper protocols for clinical trials. This can be achieved by a cooperation between the pharmaceutical industry, academia, private research centers, philanthropic institutions, and the regulatory bodies. Based on all the available information, the success of combination therapy to tackle complicated disorders such as cancer, and the blueprint already laid out on how to implement combination therapy and overcome its challenges, an argument can be made that the field has to move cautiously but quickly toward designing new clinical trials, further exploring the pathological mechanisms of AD, and re-examining the previous studies with combination therapies so that effective treatments for AD may be finally found. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
196. Brain 'metaflammasome' and risk factors associated with Alzheimer's disease
- Author
-
Taga, Mariko, Boche, Delphine, Nicoll, James, and Hugon, Jacques
- Subjects
616.8 - Abstract
A number of environmental and genetic risk factors have been identified for Alzheimer’s disease (AD) in addition to the well-known factors of ageing and APOE gene polymorphism. These include mainly lipid metabolism (e.g.diabetes) and inflammation. Recent evidence in models of the metabolic disorders obesity and type 2 diabetes suggest the involvement of a metabolic inflammasome (“metaflammasome”) in mediating chronic inflammation. The central component of the metaflammasome is the double-stranded RNA-dependent protein kinase (PKR), which also accumulates in AD brains. Other components of the metaflammasome include the proteins JNK, IKK?and IRS1. Chronic neuroinflammation is a key feature of AD. Therefore my hypothesis is that metabolic risk factors for AD activate a cerebral metaflammasome which, in turn, results in inflammation. To test my hypothesis, I investigated the expression of cerebral metaflammasome components in animals and humans. The effect of the absence of PKR on other metaflammasome components was studied using PKR-/- mice. In an acute model of systemic inflammation, using western blots and immunohistochemistry (IHC) methods, my study showed the absence of the expression of a cerebral metaflammasome in Wild-Type (WT) (n=8) and PKR-/- mice (n=3) at day 1. Similar results were obtained at day 3 (n=3 for both strains) by IHC. However, the results of our chronic high fat diet model (HFD), to reproduce obesity, showed in the WT obese mice (n=14), which developed type 2 diabetes and dyslipidaemia, increased expression of cerebral metaflammasome proteins compared to WT control mice (n=12). Interestingly, no significant difference in the expression of the metaflammasome was observed in PKR-/- HFD mice (n=5) compared to controls (n=4), supporting a key role for PKR in the metaflammasome. Furthermore, no development of type 2 diabetes and dyslipidaemia was found in PKR-/- HFD mice unlike in WT HFD mice. The inhibition of PKR and metaflammasome components could protect against metabolic disorders, which are known risk factors for AD. In the human study, analysis obtained by IHC on brain tissue from 298 participants in the Cognitive Function and Ageing Studies (CFAS) showed that high levels of PKR, JNK and IRS1 were significantly associated with poor cognitive function while a high level of IKK? was associated with good cognitive function (p < 0.05). Among participants without dementia, an increase in metaflammasome proteins was related to a lower neuropathological burden such as plaques and tangles. In contrast, among those with AD, higher levels of metaflammasome proteins were mainly associated with more severe AD pathology (p<0.05). Metaflammasome proteins were positively related to hypertension in non-demented participants, but negatively in those with AD (p<0.05). Regarding diabetes, metaflammasome proteins were negatively associated in both groups, except for JNK (p<0.05). Interestingly, in the presence of dementia, a novel relationship was observed between JNK and IKK?. APOE genotype did not affect metaflammasome proteins. In these studies, we have demonstrated for the first time the presence of a cerebral metaflammasome in the context of metabolic disorders in animals and humans. The animal data suggest an association of the cerebral metaflammasome with metabolic disorders and support the central function of PKR in the expression of metaflammasome components. The human data show an association between the metaflammasome and metabolic disorders, cognitive function and AD pathology, and highlight the modification of relationship between JNK and IKK? during dementia.
- Published
- 2015
197. Immune environment of the brain in schizophrenia and during the psychotic episode: A human post-mortem study.
- Author
-
De Picker, Livia J., Victoriano, Gerardo Mendez, Richards, Rhys, Gorvett, Alexander J., Lyons, Simeon, Buckland, George R., Tofani, Tommaso, Norman, Jeanette L., Chatelet, David S., Nicoll, James A.R., and Boche, Delphine
- Subjects
- *
GENOME-wide association studies , *TIME of death , *SCHIZOPHRENIA , *T cells , *WHITE matter (Nerve tissue) , *POSTMORTEM changes - Abstract
• Low-grade neuroinflammation is state and age-dependent in chronic schizophrenia. • 3D microglial morphology shows primed/reactive microglia in chronic schizophrenia. • Cerebral T lymphocytes recruitment consistent with a role for adaptive immunity in chronic schizophrenia. A causal relationship between immune dysregulation and schizophrenia has been supported by genome-wide association studies and epidemiological evidence. It remains unclear to what extent the brain immune environment is implicated in this hypothesis. We investigated the immunophenotype of microglia and the presence of perivascular macrophages and T lymphocytes in post-mortem brain tissue. Dorsal prefrontal cortex of 40 controls (22F:18M) and 37 (10F:27M) schizophrenia cases, of whom 16 had active psychotic symptoms at the time of death, was immunostained for seven markers of microglia (CD16, CD32a, CD64, CD68, HLA-DR, Iba1 and P2RY12), two markers for perivascular macrophages (CD163 and CD206) and T-lymphocytes (CD3). Automated quantification was blinded to the case designation and performed separately on the grey and white matter. 3D reconstruction of Iba1-positive microglia was performed in selected cases. An increased cortical expression of microglial Fcγ receptors (CD64 F = 7.92, p = 0.007; CD64/HLA-DR ratio F = 5.02, p = 0.029) highlights the importance of communication between the central and peripheral immune systems in schizophrenia. Patients in whom psychotic symptoms were present at death demonstrated an age-dependent increase of Iba1 and increased CD64/HLA-DR ratios relative to patients without psychotic symptoms. Microglia in schizophrenia demonstrated a primed/reactive morphology. A potential role for T-lymphocytes was observed, but we did not confirm the presence of recruited macrophages in the brains of schizophrenia patients. Taking in account the limitations of a post-mortem study, our findings support the hypothesis of an alteration of the brain immune environment in schizophrenia, with symptomatic state- and age-dependent effects. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
198. Microglial morphology in Alzheimer's disease and after Aβ immunotherapy.
- Author
-
Franco-Bocanegra, Diana K., Gourari, Yamina, McAuley, Ciaran, Chatelet, David S., Johnston, David A., Nicoll, James A. R., and Boche, Delphine
- Subjects
- *
ALZHEIMER'S disease , *MICROGLIA , *IMMUNOTHERAPY , *CELL motility , *CELL morphology - Abstract
Microglia are the brain immune cells and their function is highly dependent on cell motility. It was hypothesised that morphological variability leads to differences in motility, ultimately impacting on the microglial function. Here, we assessed microglial morphology in 32 controls, 44 Alzheimer's disease (AD) cases and 16 AD cases from patients immunised against Aβ42 (iAD) using 2D and 3D approaches. Our 2D assessment showed an increased number of microglia in iAD vs. AD (P = 0.032) and controls (P = 0.018). Ramified microglia were fewer in AD vs. controls (P = 0.041) but increased in iAD compared to AD (P < 0.001) and controls (P = 0.006). 3D reconstructions highlighted larger cell bodies in AD vs. controls (P = 0.049) and increased total process length in iAD vs. AD (P = 0.032), with negative correlations detected for pan-Aβ load with total process length (P < 0.001) in AD and number of primary processes (P = 0.043) in iAD. In summary, reactive/amoeboid microglia are the most represented population in the aged human brain. AD does not affect the number of microglia, but the ramified population is decreased adopting a more reactive morphology. Aβ removal by immunotherapy leads to increased ramified microglia, implying that the cells retain plasticity in an aged disease brain meriting further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
199. The role of microglia in Alzheimer's disease and following amyloid-beta-42 (Aβ42) immunisation
- Author
-
Zotova, Elina, Nicoll, James, and Boche, Delphine
- Subjects
610 ,RC0321 Neuroscience. Biological psychiatry. Neuropsychiatry - Abstract
In ammation is recognised as an important contributor to Alzheimer's disease (AD) pathogenesis alongside extracellular deposits of amyloid β protein (Aβ) and intraneuronal deposits of hyper-phosphorylated tau protein. Microglia are the key component of innate immunity within the central nervous system, and the source of in ammation in the brain. The amyloid cascade hypothesis places Aβ at a key point in the pathogenesis of AD. Consequently, immunisation against Aβ is a promising experimental treatment for AD. The current study presents a neuropathological follow-up of a unique cohort of AD patients who took part in the active Aβ42 immunisation clinical trial (AN1792, Elan) and in whom reduction in Aβ load had been observed. The hypothesis of the study is that Aβ42 immunisation affects the microglial activation level and profile associated with the reduction in Aβ. Levels of the microglial markers indicative of specific microglial functions, the microglial receptors involved in antigen recognition and uptake, complement component C1q, IgG, Aβ42 and phospho-tau in the brain tissue (frontal, temporal and parietal lobes) of immunised (n=11) and non-immunised (n=28) AD patients were analysed using immunohistochemistry. In immunised brain, a two-fold reduction in Fc receptor I (P=0.001) and Fc receptor II (P=0.002), a ten-fold reduction in macrophage scavenger receptor A (P <0.001), and a 40% reduction in lysosomal glycoprotein CD68 (P=0.018) compared to non-immunised brain were observed. Similarly, Aβ42 and phospho-tau levels were also decreased by 80% and 40% respectively. No changes in the microglial activation and antigen-presentation receptor HLA-DR, calcium-adapter channel Iba-1, or the levels of IgG and C1q were detected. The load of all microglial markers, IgG and C1q correlated with the phospho-tau load in non-immunised cases. In addition, Iba-1 and Fc RII load inversely correlated with Aβ42 load in the non-immunised group. These correlations no longer held in the immunised group. Instead, a positive correlation between the number of MSR-A positive microglial clusters and Aβ42, and an inverse correlation between C1q and Aβ42 were noted in immunised AD cases. The current study is the first of its kind in presenting a detailed immunohistochemical profile of microglia in human AD and following active Aβ42 immunisation treatment. In addition, a consistent and reproducible method for automated microscopy image analysis was developed as part of this project contributing to research methodologies. The results support the changes in the level and profile of microglial activation implied by the hypothesis. The data also suggest a link between immune system activation and the tau pathology in AD. The ability of microglia to change their profile and switch targets following stimulation by active immunisation is demonstrated in human brain.
- Published
- 2012
200. Metaflammasome components in the human brain: a role in dementia with Alzheimer's pathology?
- Author
-
Taga, Mariko, Minett, Thais, Classey, John, Matthews, Fiona E., Brayne, Carol, Ince, Paul G., Nicoll, James AR, Hugon, Jacques, and Boche, Delphine
- Subjects
- *
COGNITION disorders , *NEUROBEHAVIORAL disorders , *DEMENTIA , *BRAIN diseases , *INSULIN - Abstract
Epidemiological and genetic studies have identified metabolic disorders and inflammation as risk factors for Alzheimer's disease (AD). Evidence in obesity and type-2 diabetes suggests a role for a metabolic inflammasome ('metaflammasome') in mediating chronic inflammation in peripheral organs implicating IKKβ (inhibitor of nuclear factor kappa-B kinase subunit beta), IRS1 (insulin receptor substrate 1), JNK (c-jun N-terminal kinase), and PKR (double-stranded RNA protein kinase). We hypothesized that these proteins are expressed in the brain in response to metabolic risk factors in AD. Neocortex from 299 participants from the MRC Cognitive Function and Ageing Studies was analysed by immunohistochemistry for the expression of the phosphorylated (active) form of IKKβ [pSer176/180], IRS1 [pS312], JNK [pThr183/Tyr185] and PKR [pT451]. The data were analyzed to investigate whether the proteins were expressed together and in relation with metabolic disorders, dementia, Alzheimer's pathology and APOE genotype. We observed a change from a positive to a negative association between the proteins and hypertension according to the dementia status. Type-2 diabetes was negatively related with the proteins among participants without dementia; whereas participants with dementia and AD pathology showed a positive association with JNK. A significant association between IKKβ and JNK in participants with dementia and AD pathology was observed, but not in those without dementia. Otherwise, weak to moderate associations were observed among the protein loads. The presence of dementia was significantly associated with JNK and negatively associated with IKKβ and IRS1. Cognitive scores showed a significant positive relationship with IKKβ and a negative with IRS1, JNK and PKR. The proteins were significantly associated with pathology in Alzheimer's participants with the relationship being inverse or not significant in participants without dementia. Expression of the proteins was not related to APOE genotype. These findings highlight a role for these proteins in AD pathophysiology but not necessarily as a complex. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.