Dementia
dementia
Dr Axel Montagne
Description The Montagne lab combines molecular approaches (e.g., scRNA-Seq) with rodent noninvasive imaging, particularly magnetic resonance imaging (MRI) and two-photon microscopy, to study the causes and effects of blood-brain barrier (BBB) dysfunction in the context of neurodegenerative disease. BBB dysfunction is a major cause of inflammatory and bioenergetic deregulation in the brain, but the interplay between pericytes and endothelial cells that causes this collapse is not fully delineated. The Montagne lab is now focusing on probing BBB function and pericyte-endothelial crosstalk, especially the consequences of pericyte dysfunction on endothelial cells and the BBB, plus reciprocal signaling by activated endothelial cells. The post-holder will contribute to the development of this program by performing high-quality, original research, and producing material for publication and dissemination. Main Responsibilities: Design, perform, and analyse experiments. Master new techniques when necessary = 70 percent. Maintain accurate and up-to-date records of experimental and analytical work. Regularly summarise progress in high-quality figures. Regular reporting of progress to the PI/other members of the research team is expected. Presentation of research findings at national and international level = 10 percent. Keep up to date with related published work. Promote scientific discussion in the lab. Actively participate in the development of the lines of research and the intellectual and technical growth of the lab = 8% Take a lead role in writing up data for publication. Contribute to the dissemination and publication of own/research team’s research findings = 5 percent. To provide guidance and supervision for student projects and/or instruction of students in the use of equipment/demonstration of techniques = 4 percent. Help with the preparation of research proposals and applications to external funding bodies = 3 percent. More information and application here: https://elxw.fa.em3.oraclecloud.com/hcmUI/CandidateExperience/en/sites/CX_1001/job/1757
Dr Axel Montagne
Description The Montagne lab combines molecular approaches (e.g., scRNA-Seq) with rodent noninvasive imaging, particularly magnetic resonance imaging (MRI) and two-photon microscopy, to study the causes and effects of blood-brain barrier (BBB) dysfunction in the context of neurodegenerative disease. BBB dysfunction is a major cause of inflammatory and bioenergetic deregulation in the brain, but the interplay between pericytes and endothelial cells that causes this collapse is not fully delineated. The Montagne lab is now focusing on probing BBB function and pericyte-endothelial crosstalk, especially the consequences of pericyte dysfunction on endothelial cells and the BBB, plus reciprocal signaling by activated endothelial cells. The post-holder will contribute to the development of this program by performing high-quality, original research, and producing material for publication and dissemination. Main Responsibilities: Design, perform, and analyse experiments. Master new techniques when necessary = 70 percent. Maintain accurate and up-to-date records of experimental and analytical work. Regularly summarise progress in high-quality figures. Regular reporting of progress to the PI/other members of the research team is expected. Presentation of research findings at national and international level = 10 percent. Keep up to date with related published work. Promote scientific discussion in the lab. Actively participate in the development of the lines of research and the intellectual and technical growth of the lab = 8% Take a lead role in writing up data for publication. Contribute to the dissemination and publication of own/research team’s research findings = 5 percent. To provide guidance and supervision for student projects and/or instruction of students in the use of equipment/demonstration of techniques = 4 percent. Help with the preparation of research proposals and applications to external funding bodies = 3 percent. More information and application here: https://elxw.fa.em3.oraclecloud.com/hcmUI/CandidateExperience/en/sites/CX_1001/job/1758/share/300000300952572?utm_medium=jobshare
Nathan Skene
This project will involve the use single cell epigenetics to address key questions about the mechanisms of neurodegenerative disease. In a series of recent publications in Nature Genetics, the lab has demonstrated that single cell RNA-seq data from mice can be used to identify the cell types associated with genetic risk factors for neurodegenerative disease. To perform this mapping, we use GWAS summary statistics generated by our collaborators at a variety of international consortia and personal genomics companies. Identification of cell types genetically implicated in disease, enables better drug targeting efforts as it is free from the confounds associated with neuropathology and mouse modelling. Critical questions remain open about the etiologically relevant cell types and the regulatory effects of disease loci within those cells. We are looking to establish the use of single cell epigenetic techniques, such as ATAC-seq and CUT&TAG, as this will enhance our ability to map intergenic loci to cell type specific features. This project will involve working with human tissue brain banks, to apply these methods to targeted populations of cells. If you are interested in this position, then please email Dr Nathan Skene directly to discuss
Expanding mechanisms and therapeutic targets for neurodegenerative disease
A hallmark pathological feature of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the depletion of RNA-binding protein TDP-43 from the nucleus of neurons in the brain and spinal cord. A major function of TDP-43 is as a repressor of cryptic exon inclusion during RNA splicing. By re-analyzing RNA-sequencing datasets from human FTD/ALS brains, we discovered dozens of novel cryptic splicing events in important neuronal genes. Single nucleotide polymorphisms in UNC13A are among the strongest hits associated with FTD and ALS in human genome-wide association studies, but how those variants increase risk for disease is unknown. We discovered that TDP-43 represses a cryptic exon-splicing event in UNC13A. Loss of TDP-43 from the nucleus in human brain, neuronal cell lines and motor neurons derived from induced pluripotent stem cells resulted in the inclusion of a cryptic exon in UNC13A mRNA and reduced UNC13A protein expression. The top variants associated with FTD or ALS risk in humans are located in the intron harboring the cryptic exon, and we show that they increase UNC13A cryptic exon splicing in the face of TDP-43 dysfunction. Together, our data provide a direct functional link between one of the strongest genetic risk factors for FTD and ALS (UNC13A genetic variants), and loss of TDP-43 function. Recent analyses have revealed even further changes in TDP-43 target genes, including widespread changes in alternative polyadenylation, impacting expression of disease-relevant genes (e.g., ELP1, NEFL, and TMEM106B) and providing evidence that alternative polyadenylation is a new facet of TDP-43 pathology.
Using Fast Periodic Visual Stimulation to measure cognitive function in dementia
Fast periodic visual stimulation (FPVS) has emerged as a promising tool for assessing cognitive function in individuals with dementia. This technique leverages electroencephalography (EEG) to measure brain responses to rapidly presented visual stimuli, offering a non-invasive and objective method for evaluating a range of cognitive functions. Unlike traditional cognitive assessments, FPVS does not rely on behavioural responses, making it particularly suitable for individuals with cognitive impairment. In this talk I will highlight a series of studies that have demonstrated its ability to detect subtle deficits in recognition memory, visual processing and attention in dementia patients using EEG in the lab, at home and in clinic. The method is quick, cost-effective, and scalable, utilizing widely available EEG technology. FPVS holds significant potential as a functional biomarker for early diagnosis and monitoring of dementia, paving the way for timely interventions and improved patient outcomes.
How do we sleep?
There is no consensus on if sleep is for the brain, body or both. But the difference in how we feel following disrupted sleep or having a good night of continuous sleep is striking. Understanding how and why we sleep will likely give insights into many aspects of health. In this talk I will outline our recent work on how the prefrontal cortex can signal to the hypothalamus to regulate sleep preparatory behaviours and sleep itself, and how other brain regions, including the ventral tegmental area, respond to psychosocial stress to induce beneficial sleep. I will also outline our work on examining the function of the glymphatic system, and whether clearance of molecules from the brain is enhanced during sleep or wakefulness.
Exploring the cerebral mechanisms of acoustically-challenging speech comprehension - successes, failures and hope
Comprehending speech under acoustically challenging conditions is an everyday task that we can often execute with ease. However, accomplishing this requires the engagement of cognitive resources, such as auditory attention and working memory. The mechanisms that contribute to the robustness of speech comprehension are of substantial interest in the context of hearing mild to moderate hearing impairment, in which affected individuals typically report specific difficulties in understanding speech in background noise. Although hearing aids can help to mitigate this, they do not represent a universal solution, thus, finding alternative interventions is necessary. Given that age-related hearing loss (“presbycusis”) is inevitable, developing new approaches is all the more important in the context of aging populations. Moreover, untreated hearing loss in middle age has been identified as the most significant potentially modifiable predictor of dementia in later life. I will present research that has used a multi-methodological approach (fMRI, EEG, MEG and non-invasive brain stimulation) to try to elucidate the mechanisms that comprise the cognitive “last mile” in speech acousticallychallenging speech comprehension and to find ways to enhance them.
From the guts to the brain through adaptive immunity in the prevention of Alzheimer’ disease
Dr. Pasinetti is the Saunders Family Chair and Professor of Neurology at Icahn School of medicine at Mount Sinai, New York. His studies allowed him to develop novel therapeutic approaches through investigation of preventable risk factors including mood disorders in the promotion of resilience against neurodegenerative disorder. In his presentation Dr. Pasinetti will discuss novel concepts about the gut-brain axis in mechanisms associated to peripheral adaptive immunity as therapeutic targets to mitigate the onset and the progression of Alzheimer’s disease and other form of dementia.
Attending to the ups and downs of Lewy body dementia: An exploration of cognitive fluctuations
Dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD) share similarities in pathology and clinical presentation and come under the umbrella term of Lewy body dementias (LBD). Fluctuating cognition is a key symptom in LBD and manifests as altered levels of alertness and attention, with a marked difference between best and worst performance. Cognition and alertness can change over seconds or minutes to hours and days of obtundation. Cognitive fluctuations can have significant impacts on the quality of life of people with LBD as well as potentially contribute to the exacerbation of other transient symptoms including, for example, hallucinations and psychosis as well as making it difficult to measure cognitive effect size benefits in clinical trials of LBD. However, this significant symptom in LBD is poorly understood. In my presentation I will discuss the phenomenology of cognitive fluctuations, how we can measure it clinically and limitations of these approaches. I will then outline the work of our group and others which has been focussed on unpicking the aetiological basis of cognitive fluctuations in LBD using a variety of imaging approaches (e.g. SPECT, sMRI, fMRI and EEG). I will then briefly explore future research directions.
Diagnosing dementia using Fastball neurocognitive assessment
Fastball is a novel, fast, passive biomarker of cognitive function, that uses cheap, scalable electroencephalography (EEG) technology. It is sensitive to early dementia; language, education, effort and anxiety independent and can be used in any setting including patients’ homes. It can capture a range of cognitive functions including semantic memory, recognition memory, attention and visual function. We have shown that Fastball is sensitive to cognitive dysfunction in Alzheimer’s disease and Mild Cognitive Impairment, with data collected in patients’ homes using low-cost portable EEG. We are now preparing for significant scale-up and the validation of Fastball in primary and secondary care.
Obesity and Brain – Bidirectional Influences
The regulation of body weight relies on homeostatic mechanisms that use a combination of internal signals and external cues to initiate and terminate food intake. Homeostasis depends on intricate communication between the body and the hypothalamus involving numerous neural and hormonal signals. However, there is growing evidence that higher-level cognitive function may also influence energy balance. For instance, research has shown that BMI is consistently linked to various brain, cognitive, and personality measures, implicating executive, reward, and attentional systems. Moreover, the rise in obesity rates over the past half-century is attributed to the affordability and widespread availability of highly processed foods, a phenomenon that contradicts the idea that food intake is solely regulated by homeostasis. I will suggest that prefrontal systems involved in value computation and motivation act to limit food overconsumption when food is scarce or expensive, but promote over-eating when food is abundant, an optimum strategy from an economic standpoint. I will review the genetic and neuroscience literature on the CNS control of body weight. I will present recent studies supporting a role of prefrontal systems in weight control. I will also present contradictory evidence showing that frontal executive and cognitive findings in obesity may be a consequence not a cause of increased hunger. Finally I will review the effects of obesity on brain anatomy and function. Chronic adiposity leads to cerebrovascular dysfunction, cortical thinning, and cognitive impairment. As the most common preventable risk factor for dementia, obesity poses a significant threat to brain health. I will conclude by reviewing evidence for treatment of obesity in adults to prevent brain disease.
Exploring the Potential of High-Density Data for Neuropsychological Testing with Coregraph
Coregraph is a tool under development that allows us to collect high-density data patterns during the administration of classic neuropsychological tests such as the Trail Making Test and Clock Drawing Test. These tests are widely used to evaluate cognitive function and screen for neurodegenerative disorders, but traditional methods of data collection only yield sparse information, such as test completion time or error types. By contrast, the high-density data collected with Coregraph may contribute to a better understanding of the cognitive processes involved in executing these tests. In addition, Coregraph may potentially revolutionize the field of cognitive evaluation by aiding in the prediction of cognitive deficits and in the identification of early signs of neurodegenerative disorders such as Alzheimer's dementia. By analyzing high-density graphomotor data through techniques like manual feature engineering and machine learning, we can uncover patterns and relationships that would be otherwise hidden with traditional methods of data analysis. We are currently in the process of determining the most effective methods of feature extraction and feature analysis to develop Coregraph to its full potential.
Bridging clinical and cognitive neuroscience together to investigate semantics, above and beyond language
We will explore how neuropsychology can be leveraged to directly test cognitive neuroscience theories using the case of frontotemporal dementias affecting the language network. Specifically, we will focus on pathological, neuroimaging, and cognitive data from primary progressive aphasia. We will see how they can help us investigate the reading network, semantic knowledge organisation, and grammatical categories processing. Time permitting, the end of the talk will cover the temporal dynamics of semantic dimensions recovery and the role played by the task.
Taking the pulse of ageing: the role of cerebrovascular risk factors in ageing and dementia
Cerebrovascular support is critical for healthy cognitive ageing. Reduced cerebral blood flow in ageing is caused, among other things, by hypertension, arteriosclerosis (i.e. stiffening of the arteries) and plaque formation. Arterial stiffness is predictive of cognitive decline, is a critical risk factor for cerebrovascular accidents, and has been linked to heightened risks for Alzheimer’s Disease and other forms of dementia. The elasticity of cerebral arteries is influenced by lifestyle factors, including cardiorespiratory fitness. Monica will discuss data obtained in their laboratory with new noninvasive measures of cerebrovascular health (pulse-DOT, a diffuse optical tomographic method for studying cerebral arteriosclerosis), in conjunction with structural and functional brain measures and cognitive assessments. These findings support a model in which localised changes in arteriosclerosis lead to specific profiles of structural, functional, and cognitive declines, paving a way to individualised interventions.
Apathy and impulsivity in neurological disease – cause, effect and treatment
How do protein-RNA condensates form and contribute to disease?
In recent years, it has become clear that intrinsically disordered regions (IDRs) of RBPs, and the structure of RNAs, often contribute to the condensation of RNPs. To understand the transcriptomic features of such RNP condensates, we’ve used an improved individual nucleotide resolution CLIP protocol (iiCLIP), which produces highly sensitive and specific data, and thus enables quantitative comparisons of interactions across conditions (Lee et al., 2021). This showed how the IDR-dependent condensation properties of TDP-43 specify its RNA binding and regulatory repertoire (Hallegger et al., 2021). Moreover, we developed software for discovery and visualisation of RNA binding motifs that uncovered common binding patterns of RBPs on long multivalent RNA regions that are composed of dispersed motif clusters (Kuret et al, 2021). Finally, we used hybrid iCLIP (hiCLIP) to characterise the RNA structures mediating the assembly of Staufen RNPs across mammalian brain development, which demonstrated the roles of long-range RNA duplexes in the compaction of long 3’UTRs. I will present how the combined analysis of the characteristics of IDRs in RBPs, multivalent RNA regions and RNA structures is required to understand the formation and functions of RNP condensates, and how they change in diseases.
Human stem cell models of Alzheimer’s disease and frontotemporal dementia
The development of human induced pluripotent stem cells (iPSC) and their subsequent differentiation into neurons has provided new opportunities for the generation of physiologically-relevant, in vitro disease models. I will present our work using iPSC to modal familial Alzheimer's Disease (fAD) and Frontotemporal Dementia (FTD). We have investigated the mutation-specific effects of APP and PSEN1 mutations on Abeta generation in neurons generated from individuals with fAD, revealing distinct mechanisms that may contribute to clinical heterogeneity in disease. I will also discuss our work to understand the developmental and pathological changes to tau that occur in iPSC-neurons, particularly the challenges of understanding tau pathology in a developmental system, tau proteostasis and how iPSC-neurons may help us identify early signatures of tau pathology in disease.
Mapping Individual Trajectories of Structural and Cognitive Decline in Mild Cognitive Impairment
The US has an aging population. For the first time in US history, the number of older adults is projected to outnumber that of children by 2034. This combined with the fact that the prevalence of Alzheimer's Disease increases exponentially with age makes for a worrying combination. Mild cognitive impairment (MCI) is an intermediate stage of cognitive decline between being cognitively normal and having full-blown Dementia, with every third person with MCI progressing to dementia of the Alzheimer's Type (DAT). While there is no known way to reverse symptoms once they begin, early prediction of disease can help stall its progression and help with early financial planning. While grey matter volume loss in the Hippocampus and Entorhinal Cortex (EC) are characteristic biomarkers of DAT, little is known about the rates of decrease of these volumes within individuals in MCI state across time. We used longitudinal growth curve models to map individual trajectories of volume loss in subjects with MCI. We then looked at whether these rates of volume decrease could predict progression to DAT right in the MCI stage. Finally, we evaluated whether these rates of Hippocampal and EC volume loss were correlated with individual rates of decline of episodic memory, visuospatial ability, and executive function.
Biopsychosocial pathways in dementia inequalities
In the United States, racial/ethnic inequalities in Alzheimer's disease and related dementias persist even after controlling for socioeconomic factors and physical health. These persistent and unexplained disparities suggest: (1) there are unrecognized dementia risk factors that are socially patterned and/or (2) known dementia risk factors exhibit differential impact across social groups. Pursuing these research directions with data from multiple longitudinal studies of brain and cognitive aging has revealed several challenges to the study of late-life health inequalities, highlighted evidence for both risk and resilience within marginalized communities, and inspired new data collection efforts to advance the field.
The Role of Cerebrovascular Pathology in Aging and Neurodegenerative Disease Populations
Late-life cognitive impairment and dementia are heterogeneous and multifactorial conditions driven by a combination of genetic, vascular, and lifestyle-related factors. More than 75% of patients with dementia have evidence of cerebrovascular pathology at autopsy. Cerebrovascular disease lesions can be detected on structural MRI and used as biomarkers to determine the extent of cerebrovascular pathology. These biomarkers are associated with cognitive difficulties and increase the risk of dementia for the same level of neurodegenerative pathology. Given that some of the risk factors for cerebrovascular disease are potentially modifiable, identifying the role of cerebrovascular pathology in aging and neurodegenerative disease populations opens a window for prevention of cognitive decline and dementia.
Multimodal imaging in Dementia with Lewy bodies
Dementia with Lewy bodies (DLB) is a synucleinopathy but more than half of patients with DLB also have varying degrees of tau and amyloid-β co-pathology. Identifying and tracking the pathologic heterogeneity of DLB with multi-modal biomarkers is critical for the design of clinical trials that target each pathology early in the disease at a time when prevention or delaying the transition to dementia is possible. Furthermore, longitudinal evaluation of multi-modal biomarkers contributes to our understanding of the type and extent of the pathologic progression and serves to characterize the temporal emergence of the associated phenotypic expression. This talk will focus on the utility of multi-modal imaging in DLB.
The overlap between the spectrum of frontotemporal dementias and atypical Parkinsonism
Using vision to understand dementia in Parkinson's disease
Mechanisms to medicines in neurodegeneration
Dysregulation of protein synthesis both globally and locally in neurons and astrocytes is a key feature of neurodegenerative diseases. Aberrant signalling through the Unfolded Protein Response (UPR) and related Integrated Stress Response (ISR) have become major targets for neuroprotection in these disorders. In addition, other homeostatic mechanisms and stress responses, including the cold shock response, appear to regulate local translation and RNA splicing to control synapse maintenance and regeneration and can also be targeted therapeutically for neuroprotection. We have defined the role of UPR/ISR and the cold-shock response in neurodegenerative disorders and have developed translational strategies targeting them for new treatments for dementia.
From Vulnerable Plaque to Vulnerable Brain: Understanding the Role of Inflammation in Vascular Health, Stroke, and Cerebrovascular Disease
Every year around 100,000 people in the UK will have a stroke. Stroke is a leading cause of adult disability, and cerebrovascular disease more broadly is a major cause of dementia. Understanding these diseases – both acute and chronic manifestations of cerebrovascular disease – requires consideration not only of the brain itself, but also the blood vessels supplying it. Atherosclerosis – the hardening of arteries as we age – may predispose to stroke by triggering the formation of blood clots that block the blood supply to the brain, but also involves inflammation that may cause chronic damage to the brain and prime both the brain and body for injury. Understanding this interaction between systemic disease and brain health may have important implications for our understanding of healthy ageing and provide novel therapeutic approaches for reducing the burden of cerebrovascular disease. This talk will consider how advances in imaging may facilitate our understanding of the processes underlying atherosclerosis and how it affects the brain in stroke, as well as work currently underway to translate this understanding into improving treatments for stroke.
Neural stem cells as biomarkers of cognitive aging and dementia
Adult hippocampal neurogenesis is implicated in memory formation and mood regulation. The Thuret lab investigates environmental and molecular mechanisms controlling the production of these adult-born neurons and how they impact mental health. We study neurogenesis in healthy ageing as well as in the context of diseases such as Alzheimer’s and depression. By approaching neurogenesis in health and disease, the strategy is two folds: (i) Validating the neurogenic process as a target for prevention and pharmacological interventions. (ii) Developing neurogenesis as a biomarker of disease prediction and progression. In this talk, I will focus on presenting some recent human studies demonstrating how hippocampal neural stem cells fate can be used as biomarkers of cognitive aging and dementia.
Roles of microglia in the pathogenesis of neurodegeneration
Microglia are implicated in a variety of functions in the central nervous system, ranging from shaping neural circuits during early brain development, to surveying the brain parenchyma, and providing trophic support to neurons across the entire lifespan. In neurodegeneration, microglia have been considered for long time mere bystanders, accompanying and worsening neuronal damage. However, recent evidence indicates that microglia can causally contribute to neurodegenerative diseases, and that their dysfunction can even be at the origin of the pathology. In fact, the broad range of physiological roles microglia play in the healthy brain suggest that faulty microglia can initiate neurodegeneration through several possible mechanisms. In particular, in this seminar, we will discuss how dysfunctional microglia can affect synaptic function leading to pathological synapse loss, thus putting microglia center stage in the pathogenesis of brain disorders.
“Empowering the immune system helps defeat dementia: The key role of monocyte-derived macrophages”
Multimorbidity in the ageing human brain: lessons from neuropathological assessment
Age-associated dementias are neuropathologically characterized by the identification of hallmark intracellular and extracellular deposition of proteins, i.e., hyperphosphorylated-tau, amyloid-β, and α-synuclein, or cerebrovascular lesions. The neuropathological assessment and staging of these pathologies allows for a diagnosis of a distinct disease, e.g., amyloid-β plaques and hyperphosphorylated tau pathology in Alzheimer's disease. Neuropathological assessment in large scale cohorts, such as the UK’s Brains for Dementia Research (BDR) programme, has made it increasingly clear that the ageing brain is characterized by the presence of multiple age-associated pathologies rather than just the ‘pure’ hallmark lesion as commonly perceived. These additional pathologies can range from low/intermediate levels, that are assumed to have little if any clinical significance, to a full-blown mixed disease where there is the presence of two distinct diseases. In our recent paper (McAleese et al. 2021 Concomitant neurodegenerative pathologies contribute to the transition from mild cognitive impairment to dementia, https://alz-journals.onlinelibrary.wiley.com/doi/full/10.1002/alz.12291, Alzheimer's & Dementia), using the BDR cohort, we investigated the frequency of multimorbidity and specifically investigated the impact of additional low-level pathology on cognition. In this study, of 670 donated post-mortem brains, we found that almost 70% of cases exhibited multimorbidity and only 22% were considered a pure diagnosis. Importantly, no case of Lewy Body dementia or vascular dementia was considered pure. A key finding is that the presence of low levels of additional pathology increased the likelihood of having mild dementia vs mild cognitive impairment by almost 20-fold, indicating low levels of additional pathology do impact the clinical progression of a distinct disease. Given the high prevalence and the potential clinical impact, cerebral multimorbidity should be at the forefront of consideration in dementia research.
AI-guided solutions for early detection of neurodegenerative disorders
Despite the importance of early diagnosis of dementia for prognosis and personalised interventions, we still lack robust tools for predicting individual progression to dementia. We propose a trajectory modelling approach that mines multimodal data from patients at early dementia stages to derive individualised prognostic scores of cognitive decline Our approach has potential to facilitate effective stratification of individuals based on prognostic disease trajectories, reducing patient misclassification with important implications for clinical practice.
Mathematical models of neurodegenerative diseases
Neurodegenerative diseases such as Alzheimer’s or Parkinson’s are devastating conditions with poorly understood mechanisms and no cure. Yet, a striking feature of these conditions is the characteristic pattern of invasion throughout the brain, leading to well-codified disease stages associated with various cognitive deficits and pathologies. How can we use mathematical modelling to gain insight into this process and, doing so, gain understanding about how the brain works? In this talk, I will show that by linking new mathematical theories to recent progress in imaging, we can unravel some of the universal features associated with dementia and, more generally, brain functions.
Better Conversations - Communication Partner Training for language led dementias
Using human pluripotent stem cells to model obesity in vitro
Obesity and neurodegeneration lead to millions of premature deaths each year and lack broadly effective treatments. Obesity is largely caused by the abnormal function of cell populations in the hypothalamus that regulate appetite. We have developed methods generate human hypothalamic neurons from hPSCs to study how they respond to nutrients and hormones (e.g. leptin) and how disease-associated mutations alter their function. Since human hypothalamic neurons can be produced in large numbers, are functionally responsive, have a human genome that can be readily edited, and are in culture environment that can be readily controlled, there is an unprecedented opportunity to study the genetic and environmental factors underlying obesity. In addition, we are fascinated by the fact that mid-life obesity is a risk factor for dementia later in life, and caloric restriction, exercise, and certain anti-obesity drugs are neuroprotective, suggesting that there are shared mechanisms between obesity and neurodegeneration. Studies of HPSC-derived hypothalamic neurons may help bridge the mechanistic gulf between human genetic data and organismic phenotypes, revealing new therapeutic targets.
British Neuroscience Association (BNA) Festival - 2021
In April 2021, in partnership with the UK Dementia Research Institute, the British Neuroscience Association will host its fifth Festival of Neuroscience. Due to the ongoing uncertainty around COVID19, our 2021 event will be the first ever online Festival of Neuroscience. Although we are sorry to miss meeting in person, we're excited to create a whole new Festival experience! The ambition and scope of the BNA Festivals make them unparalleled across neuroscience. Being online will not change how the BNA2021 event will: - bring together multiple organisations with an interest in brain research at a single, shared event, creating a novel, multi-organisation forum featuring all areas of fundamental research in neuroscience and psychology, from both academia and the commercial sector, plus clinical expertise in neurology and psychiatry. - include a programme of public events as well. Past Festivals have seen a rap performance about consciousness, lunchtime talks, sessions in schools, and much more.
Challenges in Frontotemporal Dementia: clinical, genetic and pathological heterogeneity
Early constipation predicts faster dementia onset in Parkinson’s disease
Constipation is a common but not a universal feature in early PD, suggesting that gut involvement is heterogeneous and may be part of a distinct PD subtype with prognostic implications. We analysed data from the Parkinson’s Incidence Cohorts Collaboration, composed of incident community-based cohorts of PD patients assessed longitudinally over 8 years. Constipation was assessed with the MDS-UPDRS constipation item or a comparable categorical scale. Primary PD outcomes of interest were dementia, postural instability and death. PD patients were stratified according to constipation severity at diagnosis: none (n=313, 67.3%), minor (n=97, 20.9%) and major (n=55, 11.8%). Clinical progression to all 3 outcomes was more rapid in those with more severe constipation at baseline (Kaplan Meier survival analysis). Cox regression analysis, adjusting for relevant confounders, confirmed a significant relationship between constipation severity and progression to dementia, but not postural instability or death. Early constipation may predict an accelerated progression of neurodegenerative pathology. Conclusions: We show widespread cortical and subcortical grey matter micro-structure associations with schizophrenia PRS. Across all investigated phenotypes NDI, a measure of the density of myelinated axons and dendrites, showed the most robust associations with schizophrenia PRS. We interpret these results as indicative of reduced density of myelinated axons and dendritic arborization in large-scale cortico-subcortical networks mediating the genetic risk for schizophrenia.
Genetic therapies for Huntington’s disease, what does the future hold for neurodegenerative disorders?
There are no effective disease-modifying therapies for neurodegenerative diseases such as Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis or Huntington’s disease. Huntington’s disease (HD) is a devastating autosomal dominantly inherited neurodegenerative disease and the world’s most common genetic dementia. I will present an overview of important approaches in development for targeting mutant HTT DNA and RNA (Tabrizi et al Neuron 2019), the cause of HD pathogenesis, and the translational pathway from bench to clinic for a HTT targeting antisense oligonucleotide (Tabrizi et al New England Journal of Medicine 2019, Tabrizi, Science 2020) which is now in phase 3 studies. In my talk I will also review some of the genetic approaches in development for other CNS diseases. I will talk a bit about my journey as a clinician scientist and share some of my learnings for young scientists on how to survive a career in science.
CURE-ND Neurotechnology Workshop - Innovative models of neurodegenerative diseases
One of the major roadblocks to medical progress in the field of neurodegeneration is the absence of animal models that fully recapitulate features of the human diseases. Unprecedented opportunities to tackle this challenge are emerging e.g. from genome engineering and stem cell technologies, and there are intense efforts to develop models with a high translational value. Simultaneously, single-cell, multi-omics and optogenetics technologies now allow longitudinal, molecular and functional analysis of human disease processes in these models at high resolution. During this workshop, 12 experts will present recent progress in the field and discuss: - What are the most advanced disease models available to date? - Which aspects of the human disease do these accurately models, which ones do they fail to replicate? - How should models be validated? Against which reference, which standards? - What are currently the best methods to analyse these models? - What is the field still missing in terms of modelling, and of technologies to analyse disease models? CURE-ND stands for 'Catalysing a United Response in Europe to Neurodegenerative Diseases'. It is a new alliance between the German Center for Neurodegenerative Diseases (DZNE), the Paris Brain Institute (ICM), Mission Lucidity (ML, a partnership between imec, KU Leuven, UZ Leuven and VIB in Belgium) and the UK Dementia Research Institute (UK DRI). Together, these partners embrace a joint effort to accelerate the pace of scientific discovery and nurture breakthroughs in the field of neurodegenerative diseases. This Neurotechnology Workshop is the first in a series of joint events aiming at exchanging expertise, promoting scientific collaboration and building a strong community of neurodegeneration researchers in Europe and beyond.
Mapping early brain network changes in neurodegenerative and cerebrovascular disorders: a longitudinal perspective
The spatial patterning of each neurodegenerative disease relates closely to a distinct structural and functional network in the human brain. This talk will mainly describe how brain network-sensitive neuroimaging methods such as resting-state fMRI and diffusion MRI can shed light on brain network dysfunctions associated with pathology and cognitive decline from preclinical to clinical dementia. I will first present our findings from two independent datasets on how amyloid and cerebrovascular pathology influence brain functional networks cross-sectionally and longitudinally in individuals with mild cognitive impairment and dementia. Evidence on longitudinal functional network organizational changes in healthy older adults and the influence of APOE genotype will be presented. In the second part, I will describe our work on how different pathology influences brain structural network and white matter microstructure. I will also touch on some new data on how brain network integrity contributes to behavior and disease progression using multivariate or machine learning approaches. These findings underscore the importance of studying selective brain network vulnerability instead of individual region and longitudinal design. Further developed with machine learning approaches, multimodal network-specific imaging signatures will help reveal disease mechanisms and facilitate early detection, prognosis and treatment search of neuropsychiatric disorders.
Phospholipid regulation in cognitive impairment and vascular dementia
An imbalance in lipid metabolism in neurodegeneration is still poorly understood. Phospholipids (PLs) have multifactorial participation in vascular dementia as Alzheimer, post-stroke dementia, CADASIL between others. Which include the hyperactivation of phospholipases, mitochondrial stress, peroxisomal dysfunction and irregular fatty acid composition triggering proinflammation in a very early stage of cognitive impairment. The reestablishment of physiological conditions of cholesterol, sphingolipids, phospholipids and others are an interesting therapeutic target to reduce the progression of AD. We propose the positive effect of BACE1 silencing produces a balance of phospholipid profile in desaturase enzymes-depending mode to reduce the inflammation response, and recover the cognitive function in an Alzheimer´s animal and brain stroke models. Pointing out there is a great need for new well-designed research focused in preventing phospholipids imbalance, and their consequent energy metabolism impairment, pro-inflammation and enzymatic over-processing, which would help to prevent unhealthy aging and AD progression.
Targeting the synapse in Alzheimer’s Disease
Alzheimer’s Disease is characterised by the accumulation of misfolded proteins, namely amyloid and tau, however it is synapse loss which leads to the cognitive impairments associated with the disease. Many studies have focussed on single time points to determine the effects of pathology on synapses however this does not inform on the plasticity of the synapses, that is how they behave in vivo as the pathology progresses. Here we used in vivo two-photon microscopy to assess the temporal dynamics of axonal boutons and dendritic spines in mouse models of tauopathy[1] (rTg4510) and amyloidopathy[2] (J20). This revealed that pre- and post-synaptic components are differentially affected in both AD models in response to pathology. In the Tg4510 model, differences in the stability and turnover of axonal boutons and dendritic spines immediately prior to neurite degeneration was revealed. Moreover, the dystrophic neurites could be partially rescued by transgene suppression. Understanding the imbalance in the response of pre- and post-synaptic components is crucial for drug discovery studies targeting the synapse in Alzheimer’s Disease. To investigate how sub-types of synapses are affected in human tissue, the Multi-‘omics Atlas Project, a UKDRI initiative to comprehensively map the pathology in human AD, will determine the synaptome changes using imaging and synaptic proteomics in human post mortem AD tissue. The use of multiple brain regions and multiple stages of disease will enable a pseudotemporal profile of pathology and the associated synapse alterations to be determined. These data will be compared to data from preclinical models to determine the functional implications of the human findings, to better inform preclinical drug discovery studies and to develop a therapeutic strategy to target synapses in Alzheimer’s Disease[3].
Blood phosphorylated tau as biomarkers for Alzheimer’s disease
Alzheimer's disease (AD) is the most common cause of dementia, and its health and socioeconomic burdens are of major concern. Presently, a definite diagnosis of AD is established by examining brain tissue after death. These examinations focus on two major pathological hallmarks of AD in the brain: (i) amyloid plaques consisting of aggregated amyloid beta (Aβ) peptides and (ii) neurofibrillary tangles made of abnormally phosphorylated tau protein. In living individuals, AD diagnosis relies on two main approaches: (i) brain imaging of tau tangles and Aβ plaques using a technique called positron emission tomography (PET) and (ii) measuring biochemical changes in tau (including phosphorylated tau at threonine-181 [p-tau181]) and the Aβ42 peptide metabolized into CSF. Unlike Aβ42, CSF p-tau181 is highly specific for AD but its usability is restricted by the need of a lumbar puncture. Moreover, PET imaging is expensive and only available in specialised medical centres. Due to these shortcomings, a simple blood test that can detect disease-related changes in the brain is a high priority for AD research, clinical care and therapy testing. In this webinar, I will discuss the discovery of p-tau biomarkers in blood and the biochemistry of how these markers differ from those found in CSF. Furthermore, I will critically review the performance of blood p-tau biomarkers across the AD pathological process and how they associate with and predict Aβ and tau pathophysiological and neuropathological changes. Furthermore, I will evaluate the potential advantages, challenges and context of use of blood p-tau in clinical practice, therapeutic trials and population screening.
Neuron-glia interactions in synapse degeneration in Alzheimer's disease
Tara Spires-Jones’ research focuses on the mechanisms and reversibility of neurodegeneration in Alzheimer’s disease, other degenerative brain diseases, and ageing. The objective of her research group is to understand why synapses and neurons become dysfunctional and die in these diseases in order to develop effective therapeutic strategies. Her work has shown that soluble forms of the pathological proteins amyloid beta and tau contribute to synapse degeneration, and that lowering levels of these proteins can prevent and reverse phenotypes in model systems. Further, she has pioneered high-resolution imaging techniques in human post-mortem brain and found evidence that these proteins accumulate in synapses in human disease.
Multimodal brain imaging to predict progression of Alzheimer’s disease
Cross-sectional and longitudinal multimodal brain imaging studies using positron emission tomography (PET) and magnetic resonance imaging (MRI) have provided detailed insight into the pathophysiological progression of Alzheimer’s disease. It starts at an asymptomatic stage with widespread gradual accumulation of beta-amyloid and spread of pathological tau deposits. Subsequently changes of functional connectivity and glucose metabolism associated with mild cognitive impairment and brain atrophy may develop. However, the rate of progression to a symptomatic stage and ultimately dementia varies considerably between individuals. Mathematical models have been developed to describe disease progression, which may be used to identify markers that determine the current stage and likely rate of progression. Both are very important to improve the efficacy of clinical trials. In this lecture, I will provide an overview on current research and future perspectives in this area.
The interaction of sensory and motor information to shape neuronal representations in mouse cortical networks
The neurons in our brain never function in isolation; they are organized into complex circuits which perform highly specialized information processing tasks and transfer information through large neuronal networks. The aim of Janelle Pakan's research group is to better understand how neural circuits function during the transformation of information from sensory perception to behavioural output. Importantly, they also aim to further understand the cell-type specific processes that interrupt the flow of information through neural circuits in neurodegenerative disorders with dementia. The Pakan group utilizes innovative neuroanatomical tracing techniques, advanced in vivo two-photon imaging, and genetically targeted manipulations of neuronal activity to investigate the cell-type specific microcircuitry of the cerebral cortex, the macrocircuitry of cortical output to subcortical structures, and the functional circuitry underlying processes of sensory perception and motor behaviour.
Biomedical Image and Genetic Data Analysis with machine learning; applications in neurology and oncology
In this presentation I will show the opportunities and challenges of big data analytics with AI techniques in medical imaging, also in combination with genetic and clinical data. Both conventional machine learning techniques, such as radiomics for tumor characterization, and deep learning techniques for studying brain ageing and prognosis in dementia, will be addressed. Also the concept of deep imaging, a full integration of medical imaging and machine learning, will be discussed. Finally, I will address the challenges of how to successfully integrate these technologies in daily clinical workflow.
Development and Application of PET Imaging for Dementia Research
Molecular imaging using Positron Emission Tomography (PET) has become a major biomedical imaging technology. Its application towards characterisation of biochemical processes in disease could enable early detection and diagnosis, development of novel therapies and treatment evaluation. The technology is underpinned by the use of imaging probes radiolabelled with short-lived radioisotopes which can be specific and selective for biological targets in vivo e.g. markers for receptors, protein deposits, enzymes and metabolism. My talk will focus on the increasing development and application of PET imaging to clinical research in neurodegenerative diseases, for which it can be applied to delineate and understand the various pathological components of these disorders.
Population studies and ageing brains, in a time of COVID
This presentation will include a brief resume of research in older populations led from Cambridge that have informed current clinical understanding and policy regarding services and prevention for and of dementia. These population studies have more recently been ‘re-purposed’ with enthusiasm from participants into a trial platform, and this also has enabled ongoing follow-up by telephone during the COVID pandemic. Although there are no formal outputs from these latter developments general impressions will be shared.
Microglia function and dysfunction in Alzheimer’s disease
Emerging genetic studies of late-onset Alzheimer’s Disease implicate the brain’s resident macrophages in the pathogenesis of AD. More than half the risk genes associated with late-onset AD are selectively expressed in microglia and peripheral myeloid cells; yet we know little about the underlying biology or how myeloid cells contribute to AD pathogenesis. Using single-cell RNA sequencing and spatial transcriptomics we identified molecular signatures that can be used to localize and monitor distinct microglia functional states in the human and mouse brain. Our results show that microglia assume diverse functional states in development, aging and injury, including populations corresponding to known microglial functions including proliferation, migration, inflammation, and synaptic phagocytosis. We identified several innate immune pathways by which microglia recognize and prune synapses during development and in models of Alzheimer’s disease, including the classical complement cascade. Illuminating the mechanisms by which developing synaptic circuits are sculpted is providing important insight on understanding how to protect synapses in Alzheimer’s and other neurodegenerative diseases of synaptic dysfunction.
A decade of TMEM106B research
The cellular phase of Alzheimer’s Disease: from genes to cells
The amyloid cascade hypothesis for Alzheimer disease ((Hardy and Selkoe, 2002; Hardy and Higgins, 1992; Selkoe, 1991), updated in (Karran et al., 2011) provides a linear model for the pathogenesis of AD with Aβ accumulation upstream and Tau pathology, inflammation, synaptic dysfunction, neuronal loss and dementia downstream, all interlinked, initiated and driven by Aβ42 peptides or oligomers. The genetic mutations causing familial Alzheimer disease seem to support this model. The nagging problem remains however that the postulated causal, and especially the ’driving’ role of abnormal Aβ aggregation or Aβ oligomer formation could not be convincingly demonstrated until now. Indeed, many questions (e.g. what causes Aβ toxicity, what is the relation between Aβ and Tau pathology, what causes neuronal death, why is amyloid deposition not correlated with dementia etc…) were already raised when the amyloid hypothesis was conceived 25 years ago. These questions remain in essence unanswered. It seems that the old paradigm is not tenable: the amyloid cascade is too linear, too neurocentric, and does not take into account the long time lag between the biochemical phase i.e. the appearance of amyloid plaques and neuronal tangles and the ultimate clinical phase, i.e. the manifestation of dementia. The pathways linking these two phases must be complex and tortuous. We have called this the cellular phase of AD (De Strooper and Karran, 2016) to suggest that a long period of action and reaction involving neurons, neuronal circuitry but also microglia, astroglia, oligodendrocytes, and the vasculature underlies the disease. In fact it is this long disease process that should be studied in the coming years. While microglia are part of this process, they should not be considered as the only component of the cellular phase. We expect that further clinical investigations and novel tools will allow to diagnose the effects of the cellular changes in the brain and provide clinical signs for this so called preclinical or prodromal AD. Furthermore the better understanding of this phase will lead to completely novel drug targets and treatments and will lead to an era where patients will receive an appropriate therapy according to their clinical stage. In this view anti-amyloid therapy is probably only effective and useful in the very early stage of the disease and AD does no longer equal to dementia. We will discuss in our talk how single cell technology and transplantation of human iPS cells into mouse brain allow to start to map in a systematic way the cellular phase of Alzheimer’s Disease.
Epigenetics and Dementia: Lessons From the 20-Year Indianapolis-Ibadan Dementia Study
Dementia is of global interest because of the rapid increase in both the number of individuals affected and the population at risk. It is essential that the risk factors be carefully delineated for the formulation of preventive strategies. Epigenetics refers to external modifications that turn genes "on" or "off”, and cross-cultural studies of migrant populations provide information on the interplay of environmental factors on genetic predisposition. The Indianapolis-Ibadan Dementia Study compared the prevalence, incidence and risk factors of dementia in African Americans and Yoruba to tease out the role of epigenetics in dementia. The presentation will provide details on biomarkers of dementia, vascular risk factors and the association with apolipoprotein E in the Yoruba. The purpose will be to inspire early career researchers on possibilities and research strategies applicable in African populations
Semantic variant of primary progressive aphasia, clinical manifestation and underlying neuropathology
Blood Biomarkers for Neurodegenerative Dementias: An Update
More than Bystanders in Dementia, Learning What Microglia Do
Genome-wide association studies implicate microglia in Alzheimer’s disease (AD) pathogenesis, but how microglia contribute to cognitive decline in AD is unclear. Emerging research suggests microglia, the resident macrophages of the central nervous system, to be active participants in brain wiring. One mechanism by which microglia help eliminate synapses is through the classical complement pathway (C1q, CR3/C3). Data from multiple laboratories collectively suggest that there may be an aberrant reactivation of the complement-dependent pruning pathway in multiple models of neurologic diseases including AD. These data altogether suggest that microglia participate in synaptic pathology. However, how and which synapses are targeted are unknown. Furthermore, whether microglia directly impair synaptic function is unknown. Primary goals of my laboratory are to understand how higher cognitive functions such as learning and memory involve microglial biology in the healthy adult brain and dissect immune mechanisms behind the region-specific vulnerability of synapse loss and neuronal dysfunction during disease. Mechanistic insight into local signals that regulate neuroglia interactions will be key to developing potential therapeutic avenues to target in disease.
Mechanisms of pathogenesis in the tauopathies
The distribution of pathological tau in the brain of patients with AD is highly predicable, and as disease worsens, it spreads transynaptically from initial regions of vulnerability. The reason why only some neurons are vulnerable to the accumulation and propagation of pathological forms of tau, and the mechanisms by which tauopathy spreads through the brain are not well understood. Using a combination of immunohistochemistry and computational analysis we have examined pathway differences between vulnerable and resistant neurons. How tau spreads across a synapse has been examined in vitro using different model systems. Our data show that dysregulation of tau homeostasis determines the cellular and regional vulnerability of specific neurons to tau pathology (H. Fu et al. 2019. Nat. Neuro. 22 (1):47-56) and that deficits in tau homeostasis can exacerbate tau accumulation and propagation. Aging appears to impact similar neuronal populations. Mechanisms and consequences of abnormal tau accumulation within neurons, its transfer between cells, pathology propagation and therapeutic opportunities will be discussed.
Novel immunotherapy to treat Alzheimer’s disease and Dementia: from curiosity-driven research to prospect of therapy
Accumulation of phospho-alpha synuclein and oligomeric tau in presynapses in Parkinson’s disease and Dementia with Lewy Bodies
FENS Forum 2024
Coactivation pattern analysis reveals changes in prodromal dementia with Lewy bodies
FENS Forum 2024
A comparison of the brain parameters from Alzheimer’s and non-Alzheimer’s dementia in Thais: A pilot study
FENS Forum 2024
Development of a novel viral vector-based model of dementia with Lewy bodies in mice
FENS Forum 2024
Early hippocampal hyperexcitability and mitochondrial changes in a transgenic mouse model of dementia with Lewy bodies
FENS Forum 2024
Exploring the combinatorial, diagnostic utility of multimodal biomarkers in differentially diagnosing Dementia with Lewy Bodies from Alzheimer’s through predictive statistical modelling
FENS Forum 2024
Identification of sex-specific autophagy enhancers for dementia
FENS Forum 2024
Influence of the APOE4 risk factor on hippocampal epigenetic and transcriptomic signatures in a physiological and pathological environment indicative of dementia with Lewy bodies (DLB)
FENS Forum 2024
The influence of depression onset timing after gastrointestinal disease on dementia risk
FENS Forum 2024
Investigating the association between the novel GAP-43 concentration with diffusion tensor imaging indices in Alzheimer's dementia continuum
FENS Forum 2024
Investigating the molecular basis for selective vulnerability in FET-linked amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD)
FENS Forum 2024
Irisin levels in cerebrospinal fluid correlate with biomarkers and clinical dementia scores in Alzheimer’s disease
FENS Forum 2024
miR-302 induced by glucagon-like peptide-1 (GLP-1) signaling reduces the α-synuclein neurotoxicity linked to dementia with Lewy bodies (DLB)
FENS Forum 2024
Molecular elucidation of lipofuscin in the aging brain and a rare pediatric dementia
FENS Forum 2024
Serum irisin levels positively correlate with neuropsychological scores in patients with dementia and could be a potential biomarker of Alzheimer’s disease
FENS Forum 2024
Synaptic gene expression changes in frontotemporal dementia due to the MAPT 10+16 mutation
FENS Forum 2024
Synergistic effects of α-synuclein, tau, and amyloid pathology on mitophagy in dementia with Lewy bodies
FENS Forum 2024
Transcranial pulse stimulation for the treatment of patients with Alzheimer's dementia
FENS Forum 2024
Unraveling the impact of tau on retinal development: Insights from human iPSC-derived models of frontotemporal dementia (FTD)
FENS Forum 2024
Validating a data-driven EEG-based dementia screening test
FENS Forum 2024