Biomarkers
biomarkers
Prof. Pierre Mégevand
The Human Neuron Lab (@LabNeuron), led by Prof. Pierre Mégevand, is dedicated to advancing the detection and prediction of epileptic seizures. The lab also investigates the neuronal basis of human cognitive brain functions. For that purpose, the lab focuses on invasive neurophysiology in the human brain, including ECoG and stereo-EEG. Additionally, unique microelectrode recordings (using Utah arrays and microwire electrodes) give access to the activity of dozens of single neurons in the patient's brain in order to reveal novel markers of epileptic seizures at the neuronal population level. The lab is equipped with state-of-the-art technology for human invasive neurophysiology. It benefits from the powerful computing infrastructure of the University. Importantly, the lab is fully integrated with the epilepsy monitoring unit of Geneva University Hospitals, and thus boasts exceptional access to patients and recordings. This project focuses on defining novel markers of seizures in patients who suffer from epilepsy. Continuous intracranial EEG and microelectrode recordings will be acquired for several weeks. Single-unit activity will be tracked over time for multiple neurons. Activity within the neuronal population will be examined for the presence of patterns that are specific to the patient’s seizures. The performance of seizure detection and prediction using microelectrode recordings will be compared to existing algorithms based on intracranial EEG data. Research tasks: - Acquire, analyze, and curate a uniquely rich dataset of human intracranial EEG and microelectrode recordings - Build a pipeline for semi-automated single-neuron identification and tracking - Establish novel markers of neuronal population activity that identify seizures - Participate in the mapping of sensory, motor and language functions in epilepsy patients - Daily interactions with the patients and staff of the epilepsy monitoring unit Work environment: The University of Geneva is a prestigious research hub in neuroscience, federating many labs that cover the full spectrum from basic to cognitive, translational and clinical research. The neuroscience community in Geneva is also strengthened by rich collaborations with other research institutions, including Campus Biotech, the Wyss Center, and the EPFL. This project is fully funded by a grant from the Swiss National Science Foundation. The PhD and post-doc positions are open for up to 4 years each. Swiss salaries are very attractive in international comparison. The positions will open from May 2021 onwards. Please send your application, including a letter of intent, curriculum vitae, list of publications, and at least two references, by e-mail to: Prof. Pierre Mégevand Division of neurology, Geneva University Hospitals Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland pierre.megevand@unige.ch
Dr. Stéphane Maison
A NIH-funded postdoctoral position is immediately available in Dr. Stéphane Maison’s laboratory in the Department of Otolaryngology – Head & Neck Surgery at the Massachusetts Eye & Ear – Harvard Medical School. Our research interests focus on identifying biomarkers of a large range of etiologies and their associated disorders including difficulties hearing and communicating in noisy environments, reduced sound level tolerance and tinnitus using a test battery based on behavioral, electrophysiologic, and psychophysical measures. Salary and benefits are consistent with NIH guidelines and institution policies based on applicant’s experience. Highly motivated candidates who recently graduated with a PhD in biomedical engineering, computational biology, hearing science, neuroscience, or other related fields are welcome to apply. The applicant should have strong programming skills (e.g., Matlab, Python), independent and productive. Experience with human testing is preferred but not required. The fellow will receive an appointment at Massachusetts Eye and Ear and Harvard Medical School. Interested applicants should apply using the following link: https://partners.taleo.net/careersection/mee/jobdetail.ftl?job=3299643&tz=GMT-04%3A00&tzname=America%2FNew_York
Axel Hutt
The National Institute for Computer Science and Control (INRIA) provides a postdoctoral fellowship on Mathematical modelling of neuronal EEG activity under brain stimulation. We are interested in developing neurostimulation techniques in order to improve the cure of patients suffering from mental disorders. To this end, our aim is to develop dynamic neural models and merging these data to experimentally observed data, such as EEG or BOLD responses. This merge may utilize diverse optimization techniques, such as data assimilation. The latter permits to estimate model parameters adaptively in non-stationary signals, i.e. online in time. A prominent example for a data assimilation technique is Kalman filtering. More detailed, we are looking for collaborators, who are interested in neural population models describing macroscopic brain activity in pathological brain states under neurostimulation. The mathematical analysis of such models typically yields important insights into the origin of the brain activity. Moreover, the merge with experimental data demands a certain understanding of data analysis techniques to prepare the experimental data and identify correctly good biomarkers. It would be advantageous if the candidate has some fundamental expertise in this respect. Finally, the perfect future collaborator has already some expertise in parameter estimation techniques, especially in data assimilation.
SRF | Dreem EEG Headband to Assess Sleep EEG Biomarkers in SYNGAP1
Personalized medicine and predictive health and wellness: Adding the chemical component
Wearable sensors that detect and quantify biomarkers in retrievable biofluids (e.g., interstitial fluid, sweat, tears) provide information on human dynamic physiological and psychological states. This information can transform health and wellness by providing actionable feedback. Due to outdated and insufficiently sensitive technologies, current on-body sensing systems have capabilities limited to pH, and a few high-concentration electrolytes, metabolites, and nutrients. As such, wearable sensing systems cannot detect key low-concentration biomarkers indicative of stress, inflammation, metabolic, and reproductive status. We are revolutionizing sensing. Our electronic biosensors detect virtually any signaling molecule or metabolite at ultra-low levels. We have monitored serotonin, dopamine, cortisol, phenylalanine, estradiol, progesterone, and glucose in blood, sweat, interstitial fluid, and tears. The sensors are based on modern nanoscale semiconductor transistors that are straightforwardly scalable for manufacturing. We are developing sensors for >40 biomarkers for personalized continuous monitoring (e.g., smartwatch, wearable patch) that will provide feedback for treating chronic health conditions (e.g., perimenopause, stress disorders, phenylketonuria). Moreover, our sensors will enable female fertility monitoring and the adoption of more healthy lifestyles to prevent disease and improve physical and cognitive performance.
Applied cognitive neuroscience to improve learning and therapeutics
Advancements in cognitive neuroscience have provided profound insights into the workings of the human brain and the methods used offer opportunities to enhance performance, cognition, and mental health. Drawing upon interdisciplinary collaborations in the University of California San Diego, Human Performance Optimization Lab, this talk explores the application of cognitive neuroscience principles in three domains to improve human performance and alleviate mental health challenges. The first section will discuss studies addressing the role of vision and oculomotor function in athletic performance and the potential to train these foundational abilities to improve performance and sports outcomes. The second domain considers the use of electrophysiological measurements of the brain and heart to detect, and possibly predict, errors in manual performance, as shown in a series of studies with surgeons as they perform robot-assisted surgery. Lastly, findings from clinical trials testing personalized interventional treatments for mood disorders will be discussed in which the temporal and spatial parameters of transcranial magnetic stimulation (TMS) are individualized to test if personalization improves treatment response and can be used as predictive biomarkers to guide treatment selection. Together, these translational studies use the measurement tools and constructs of cognitive neuroscience to improve human performance and well-being.
From rare Genetic cohorts of Parkinsonism to biomarkers and to understanding broader neurodegenerative disease mechanisms
Quasicriticality and the quest for a framework of neuronal dynamics
Critical phenomena abound in nature, from forest fires and earthquakes to avalanches in sand and neuronal activity. Since the 2003 publication by Beggs & Plenz on neuronal avalanches, a growing body of work suggests that the brain homeostatically regulates itself to operate near a critical point where information processing is optimal. At this critical point, incoming activity is neither amplified (supercritical) nor damped (subcritical), but approximately preserved as it passes through neural networks. Departures from the critical point have been associated with conditions of poor neurological health like epilepsy, Alzheimer's disease, and depression. One complication that arises from this picture is that the critical point assumes no external input. But, biological neural networks are constantly bombarded by external input. How is then the brain able to homeostatically adapt near the critical point? We’ll see that the theory of quasicriticality, an organizing principle for brain dynamics, can account for this paradoxical situation. As external stimuli drive the cortex, quasicriticality predicts a departure from criticality while maintaining optimal properties for information transmission. We’ll see that simulations and experimental data confirm these predictions and describe new ones that could be tested soon. More importantly, we will see how this organizing principle could help in the search for biomarkers that could soon be tested in clinical studies.
From cells to systems: multiscale studies of the epileptic brain
It is increasingly recognized that epilepsy affects human brain organization across multiple scales, ranging from cellular alterations in specific regions towards macroscale network imbalances. My talk will overview an emerging paradigm that integrates cellular, neuroimaging, and network modelling approaches to faithful characterize the extent of structural and functional alterations in the common epilepsies. I will also discuss how multiscale framework can help to derive clinically useful biomarkers of dysfunction, and how these methods may guide surgical planning and prognostics.
Integration of 3D human stem cell models derived from post-mortem tissue and statistical genomics to guide schizophrenia therapeutic development
Schizophrenia is a neuropsychiatric disorder characterized by positive symptoms (such as hallucinations and delusions), negative symptoms (such as avolition and withdrawal) and cognitive dysfunction1. Schizophrenia is highly heritable, and genetic studies are playing a pivotal role in identifying potential biomarkers and causal disease mechanisms with the hope of informing new treatments. Genome-wide association studies (GWAS) identified nearly 270 loci with a high statistical association with schizophrenia risk; however each locus confers only a small increase in risk therefore it is difficult to translate these findings into understanding disease biology that can lead to treatments. Induced pluripotent stem cell (iPSC) models are a tractable system to translate genetic findings and interrogate mechanisms of pathogenesis. Mounting research with patient-derived iPSCs has proposed several neurodevelopmental pathways altered in SCZ, such as neural progenitor cell (NPC) proliferation, imbalanced differentiation of excitatory and inhibitory cortical neurons. However, it is unclear what exactly these iPS models recapitulate, how potential perturbations of early brain development translates into illness in adults and how iPS models that represent fetal stages can be utilized to further drug development efforts to treat adult illness. I will present the largest transcriptome analysis of post-mortem caudate nucleus in schizophrenia where we discovered that decreased presynaptic DRD2 autoregulation is the causal dopamine risk factor for schizophrenia (Benjamin et al, Nature Neuroscience 2022 https://doi.org/10.1038/s41593-022-01182-7). We developed stem cell models from a subset of the postmortem cohort to better understand the molecular underpinnings of human psychiatric disorders (Sawada et al, Stem Cell Research 2020). We established a method for the differentiation of iPS cells into ventral forebrain organoids and performed single cell RNAseq and cellular phenotyping. To our knowledge, this is the first study to evaluate iPSC models of SZ from the same individuals with postmortem tissue. Our study establishes that striatal neurons in the patients with SCZ carry abnormalities that originated during early brain development. Differentiation of inhibitory neurons is accelerated whereas excitatory neuronal development is delayed, implicating an excitation and inhibition (E-I) imbalance during early brain development in SCZ. We found a significant overlap of genes upregulated in the inhibitory neurons in SCZ organoids with upregulated genes in postmortem caudate tissues from patients with SCZ compared with control individuals, including the donors of our iPS cell cohort. Altogether, we demonstrate that ventral forebrain organoids derived from postmortem tissue of individuals with schizophrenia recapitulate perturbed striatal gene expression dynamics of the donors’ brains (Sawada et al, biorxiv 2022 https://doi.org/10.1101/2022.05.26.493589).
Integrative Neuromodulation: from biomarker identification to optimizing neuromodulation
Why do we make decisions impulsively blinded in an emotionally rash moment? Or caught in the same repetitive suboptimal loop, avoiding fears or rushing headlong towards illusory rewards? These cognitive constructs underlying self-control and compulsive behaviours and their influence by emotion or incentives are relevant dimensionally across healthy individuals and hijacked across disorders of addiction, compulsivity and mood. My lab focuses on identifying theory-driven modifiable biomarkers focusing on these cognitive constructs with the ultimate goal to optimize and develop novel means of neuromodulation. Here I will provide a few examples of my group’s recent work to illustrate this approach. I describe a series of recent studies on intracranial physiology and acute stimulation focusing on risk taking and emotional processing. This talk highlights the subthalamic nucleus, a common target for deep brain stimulation for Parkinson’s disease and obsessive-compulsive disorder. I further describe recent translational work in non-invasive neuromodulation. Together these examples illustrate the approach of the lab highlighting modifiable biomarkers and optimizing neuromodulation.
Development of Interictal Networks: Implications for Epilepsy Progression and Cognition
Epilepsy is a common and disabling neurologic condition affecting adults and children that results from complex dysfunction of neural networks and is ineffectively treated with current therapies in up to one third of patients. This dysfunction can have especially severe consequences in pediatric age group, where neurodevelopment may be irreversibly affected. Furthermore, although seizures are the most obvious manifestation of epilepsy, the cognitive and psychiatric dysfunction that often coexists in patients with this disorder has the potential to be equally disabling. Given these challenges, her research program aims to better understand how epileptic activity disrupts the proper development and function of neural networks, with the overall goal of identifying novel biomarkers and systems level treatments for epileptic disorders and their comorbidities, especially those affecting children.
Baby steps to breakthroughs in precision health in neurodevelopmental disorders
The glymphatic system in motor neurone disease
Neurodegenerative diseases are chronic and inexorable conditions characterised by the presence of insoluble aggregates of abnormally ubiquinated and phosphorylated proteins. Recent evidence also suggests that protein misfolding can propagate throughout the body in a prion-like fashion via the interstitial or cerebrospinal fluids (CSF). As protein aggregation occurs well before the onset of brain damage and symptoms, new biomarkers sensitive to early pathology, together with therapeutic strategies that include eliminating seed proteins and blocking cell-to-cell spread, are of vital importance. The glymphatic system, which facilitates the continuous exchange of CSF and interstitial fluid to clear the brain of waste, presents as a potential biomarker of disease severity, therapeutic target, and drug delivery system. In this webinar, Associate Professor David Wright from the Department of Neuroscience, Monash University, will outline recent advances in using MRI to investigate the glymphatic system. He will also present some of his lab’s recent work investigating glymphatic clearance in preclinical models of motor neurone disease. Associate Professor David Wright is an NHMRC Emerging Leadership Fellow and the Director of Preclinical Imaging in the Department of Neuroscience, Monash University and the Alfred Research Alliance, Alfred Health. His research encompasses the development, application and analysis of advanced magnetic resonance imaging techniques for the study of disease, with a particular emphasis on neurodegenerative disorders. Although less than three years post PhD, he has published over 60 peer-reviewed journal articles in leading neuroscience journals such as Nature Medicine, Brain, and Cerebral Cortex.
Biofluid biomarkers in atypical parkinsonism
PET imaging in brain diseases
Talk 1. PET based biomarkers of treatment efficacy in temporal lobe epilepsy A critical aspect of drug development involves identifying robust biomarkers of treatment response for use as surrogate endpoints in clinical trials. However, these biomarkers also have the capacity to inform mechanisms of disease pathogenesis and therapeutic efficacy. In this webinar, Dr Bianca Jupp will report on a series of studies using the GABAA PET ligand, [18F]-Flumazenil, to establish biomarkers of treatment response to a novel therapeutic for temporal lobe epilepsy, identifying affinity at this receptor as a key predictor of treatment outcome. Dr Bianca Jupp is a Research Fellow in the Department of Neuroscience, Monash University and Lead PET/CT Scientist at the Alfred Research Alliance–Monash Biomedical Imaging facility. Her research focuses on neuroimaging and its capacity to inform the neurobiology underlying neurological and neuropsychiatric disorders. Talk 2. The development of a PET radiotracer for reparative microglia Imaging of neuroinflammation is currently hindered by the technical limitations associated with TSPO imaging. In this webinar, Dr Lucy Vivash will discuss the development of PET radiotracers that specifically image reparative microglia through targeting the receptor kinase MerTK. This includes medicinal chemistry design and testing, radiochemistry, and in vitro and in vivo testing of lead tracers. Dr Lucy Vivash is a Research Fellow in the Department of Neuroscience, Monash University. Her research focuses on the preclinical development and clinical translation of novel PET radiotracers for the imaging of neurodegenerative diseases.
Angelman syndrome: biomarkers and treatment opportunities
ISYNC: International SynAGE Conference on Healthy Ageing
The SynAGE committee members are thrilled to host ISYNC, the International SynAGE conference on healthy ageing, on 28-30 March 2022 in Magdeburg, Germany. This conference has been entirely organised from young scientists of the SynAGE research training group RTG 2413 (www.synage.de) and represents a unique occasion for researchers from all over the world to bring together and join great talks and sessions with us and our guests. A constantly updated list of our speakers can be found on the conference webpage: www.isync-md.de. During the conference, attendees will have access to a range of symposia which will deal with Glia, Biomarkers and Immunoresponses during ageing to neurodegeneration brain integrity and cognitive function in health and diseases. Moreover, the conference will offer social events especially for young researchers and the possibility to network together in a beautiful and suggestive location where our conference will take place: the Johanniskirche. The event will be happening in person, but due to the current pandemic situation and restrictions we are planning the conference as a hybrid event with lots of technical support to ensure that every participant can follow the talks and take part in the scientific discussions. The registration to our ISYNC conference is free of charge. However, the number of people attending the conference in person is restricted to 100. Afterwards, registrations will be accepted for joining virtually only. The registration is open until 15.02.2022. Especially for PhD and MD Students: Check our available Travel Grants, Poster Prize and SynAGE Award Dinner: https://www.isync-md.de/index.php/phd-md-specials/ If you need any further information don’t hesitate to contact us via email: contact@synage.de. We are looking forward to meet you in 2022 in Magdeburg to discuss about our research and ideas and bless together science. Your ISYNC organization Committee
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.
Multi-modal biomarkers improve prediction of memory function in cognitively unimpaired older adults
Identifying biomarkers that predict current and future cognition may improve estimates of Alzheimer’s disease risk among cognitively unimpaired older adults (CU). In vivo measures of amyloid and tau protein burden and task-based functional MRI measures of core memory mechanisms, such as the strength of cortical reinstatement during remembering, have each been linked to individual differences in memory in CU. This study assesses whether combining CSF biomarkers with fMRI indices of cortical reinstatement improves estimation of memory function in CU, assayed using three unique tests of hippocampal-dependent memory. Participants were 158 CU (90F, aged 60-88 years, CDR=0) enrolled in the Stanford Aging and Memory Study (SAMS). Cortical reinstatement was quantified using multivoxel pattern analysis of fMRI data collected during completion of a paired associate cued recall task. Memory was assayed by associative cued recall, a delayed recall composite, and a mnemonic discrimination task that involved discrimination between studied ‘target’ objects, novel ‘foil’ objects, and perceptually similar ‘lure’ objects. CSF Aβ42, Aβ40, and p-tau181 were measured with the automated Lumipulse G system (N=115). Regression analyses examined cross-sectional relationships between memory performance in each task and a) the strength of cortical reinstatement in the Default Network (comprised of posterior medial, medial frontal, and lateral parietal regions) during associative cued recall and b) CSF Aβ42/Aβ40 and p-tau181, controlling for age, sex, and education. For mnemonic discrimination, linear mixed effects models were used to examine the relationship between discrimination (d’) and each predictor as a function of target-lure similarity. Stronger cortical reinstatement was associated with better performance across all three memory assays. Age and higher CSF p-tau181 were each associated with poorer associative memory and a diminished improvement in mnemonic discrimination as target-lure similarity decreased. When combined in a single model, CSF p-tau181 and Default Network reinstatement strength, but not age, explained unique variance in associative memory and mnemonic discrimination performance, outperforming the single-modality models. Combining fMRI measures of core memory functions with protein biomarkers of Alzheimer’s disease significantly improved prediction of individual differences in memory performance in CU. Leveraging multimodal biomarkers may enhance future prediction of risk for cognitive decline.
Emerging Treatment Options in Psychiatry
The World Health Organization (WHO) estimates that untreated mental disorders accountfor 13% of the total global burden of disease, and by 2030, depression alone will be the leadingcause of disability around the world – outpacing heart disease, cancer, and HIV. This grim pictureis further compounded by the mental health burden delivered by the coronavirus pandemic.The lack of novel treatment options in psychiatry is restricted by a limited understanding in theneuroscience basis of mental disorders, availability of relevant biomarkers, poor predictability inanimal models, and high failure rates in psychiatric drug development. However, theannouncement in 2019 from the Federal Drug Administration (FDA) for approvals of newinterventions for treatment-resistant depression (intranasal esketamine) and postpartumdepression (i.v. brexanolone), demand critical attention. Novel public-private partnerships indrug discovery, new translational data on co-morbid biology, in particular the ascendance ofpsycho-immunology, have highlighted the arrival of a new frontier in biological psychiatryresearch for depressive disorders.
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.
Translational Biomarkers in Preclinical Models of Neurodevelopmental Disorders
Evidence for the role of glymphatic dysfunction in the development of Alzheimer’s disease
Glymphatic perivascular exchange is supported by the astroglial water channel aquaporin-4 (AQP4), which localizes to perivascular astrocytic endfeet surrounding the cerebral vasculature. In aging mice, impairment of glymphatic function is associated with reduced perivascular AQP4 localization, yet whether these changes contribute to the development of neurodegenerative disease, such as Alzheimer’s disease (AD), remains unknown. Using post mortem human tissue, we evaluated perivascular AQP4 localization in the frontal cortical gray matter, white matter, and hippocampus of cognitively normal subjects and those with AD. Loss of perivascular and increasing cellular localization of AQP4 in the frontal gray matter was specifically associated with AD status, amyloid β (Aβ) and tau pathology, and cognitive decline in the early stages of disease. Using AAV-PHP.B to drive expression on non-perivascular AQP4 in wild type and Tg2576 (APPSwe, mouse model of Aβ deposition) mice, increased cellular AQP4 localization did not slow glymphatic function or change Aβ deposition. Using the Snta1 knockout line (which lacks perivascular AQP4 localization), we observed that loss AQP4 from perivascular endfeet slowed glymphatic function in wild type mice and accelerated Aβ plaque deposition in Tg2576 mice. These findings demonstrate that loss of perivascular AQP4 localization, and not increased cellular AQP4 localization, slows glymphatic function and promotes the development of AD pathology. To evaluate whether naturally occurring variation in the human AQP4 gene, or the alpha syntrophin (SNTA1), dystrobrevin (DTNA) or dystroglycan (DAG1) genes (whose products maintain perivascular AQP4 localization) confer risk for or protection from AD pathology or clinical progression, we evaluated 56 tag single nucleotide polymorphisms (SNPs) across these genes for association with CSF AD biomarkers, MRI measures of cortical and hippocampal atrophy, and longitudinal cognitive decline in the Alzheimer’s Disease Neuroimaging Initiative I (ADNI I) cohort. We identify 25 different significant associations between AQP4, SNTA1, DTNA, and DAG1 tag SNPs and phenotypic measures of AD pathology and progression. These findings provide complimentary human genetic evidence for the contribution of perivascular glymphatic dysfunction to the development of AD in human populations.
Dancing to a Different Tune: TANGO Gives Hope for Dravet Syndrome
The long-term goal of our research is to understand the mechanisms of SUDEP, defined as Sudden, Unexpected, witnessed or unwitnessed, nontraumatic and non-drowning Death in patients with EPilepsy, excluding cases of documented status epilepticus. The majority of SUDEP patients die during sleep. SUDEP is the most devastating consequence of epilepsy, yet little is understood about its causes and no biomarkers exist to identify at risk patients. While SUDEP accounts for 7.5-20% of all epilepsy deaths, SUDEP risk in the genetic epilepsies varies with affected genes. Patients with ion channel gene variants have the highest SUDEP risk. Indirect evidence variably links SUDEP to seizure-induced apnea, pulmonary edema, dysregulation of cerebral circulation, autonomic dysfunction, and cardiac arrhythmias. Arrhythmias may be primary or secondary to hormonal or metabolic changes, or autonomic discharges. When SUDEP is compared to Sudden Cardiac Death secondary to Long QT Syndrome, especially to LQT3 linked to variants in the voltage-gated sodium channel (VGSC) gene SCN5A, there are parallels in the circumstances of death. To gain insight into SUDEP mechanisms, our approach has focused on channelopathies with high SUDEP incidence. One such disorder is Dravet syndrome (DS), a devastating form of developmental and epileptic encephalopathy (DEE) characterized by multiple pharmacoresistant seizure types, intellectual disability, ataxia, and increased mortality. While all patients with epilepsy are at risk for SUDEP, DS patients may have the highest risk, up to 20%, with a mean age at SUDEP of 4.6 years. Over 80% of DS is caused by de novo heterozygous loss-of-function (LOF) variants in SCN1A, encoding the VGSC Nav1.1 subunit, resulting in haploinsufficiency. A smaller cohort of patients with DS or a more severe DEE have inherited, homozygous LOF variants in SCN1B, encoding the VGSC 1/1B non-pore-forming subunits. A related DEE, Early Infantile EE (EIEE) type 13, is linked to de novo heterozygous gain-of-function variants in SCN8A, encoding the VGSC Nav1.6. VGSCs underlie the rising phase and propagation of action potentials in neurons and cardiac myocytes. SCN1A, SCN8A, and SCN1B are expressed in both the heart and brain of humans and mice. Because of this, we proposed that cardiac arrhythmias contribute to the mechanism of SUDEP in DEE. We have taken a novel approach to the development of therapeutics for DS in collaboration with Stoke Therapeutics. We employed Targeted Augmentation of Nuclear Gene Output (TANGO) technology, which modulates naturally occurring, non-productive splicing events to increase target gene and protein expression and ameliorate disease phenotype in a mouse model. We identified antisense oligonucleotides (ASOs) that specifically increase the expression of productive Scn1a transcript in human and mouse cell lines, as well as in mouse brain. We showed that a single intracerebroventricular dose of a lead ASO at postnatal day 2 or 14 reduced the incidence of electrographic seizures and SUDEP in the F1:129S-Scn1a+/- x C57BL/6J mouse model of DS. Increased expression of productive Scn1a transcript and NaV1.1 protein were confirmed in brains of treated mice. Our results suggest that TANGO may provide a unique, gene-specific approach for the treatment of DS.
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.
From epigenetics to stratified therapies in neuropsychiatric diseases
The establishment of effective therapies for neurodegenerative and neuropsychiatric diseases is still challenging and one of the reasons is that especially for age-associated neurodegenerative diseases pathology accumulates long before there are any clinical signs of disease. Thus, patients are often only diagnosed at an already advanced state of molecular pathology, when causative therapies fail. Thus, there is an urgent need for molecular biomarkers that could detect individuals at risk for developing a CNS disease and stratify patients. I will address epigenetic processes such as histone-modifications and non-coding RNAs as potential approaches for patient stratification and therapeutic interaction, with a specific focus on RNA-therapies. Here, I plan to cover examples from our recent research on Alzheimer’s disease and Schizophrenia.
The pathophysiology of prodromal Parkinson’s disease
Studying the pathophysiology of late stage Parkinson’s disease (PD) – after the patients have experienced severe neuronal loss – has helped develop various symptomatic treatments for PD (e.g., deep brain stimulation). However, it has been of limited use in developing neuroprotective disease-modifying therapies (DMTs), because DMTs require interventions at much earlier stages of PD when vulnerable neurons are still intact. Because PD patients exhibit various non-motor prodromal symptoms (ie, symptoms that predate diagnosis), understanding the pathophysiology underlying these symptom could lead to earlier diagnosis and intervention. In my talk, I will present a recently elucidated example of how PD pathologies alter the channel biophysics of intact vagal motoneurons (known to be selectively vulnerable in PD) to drive dysautonomia that is reminiscent of prodromal PD. I will discuss how elucidating the pathophysiology of prodromal symptoms can lead to earlier diagnosis through the development of physiological biomarkers for PD.
Hughlings Jackson Lecture: Making Progress in Progressive MS – the Ultimate Challenge!
On April 22, 2021, Dr. Alan J Thompson of the University College London and the UCL Institute of Neurology, London, UK will deliver the Hughlings Jackson Lecture entitled, “Making Progress in Progressive MS – the Ultimate Challenge!” Established in 1935, the Hughlings Jackson Lecture is The Neuro’s premier scientific lecture. It honors the legacy of British neurologist John Hughlings Jackson (1835-1911) who pioneered the development of neurology as a medical specialty. Talk Abstract : The international focus on progressive MS, driven by the Progressive MS Alliance amongst others, together with recent encouraging results from clinical trials have raised the profile and emphasised the importance of understanding, treating and ultimately preventing progression in MS. Effective treatment for Progressive MS is now regarded as the single most important issue facing the MS community. There are several important challenges to developing new treatments for progressive MS. Fundamental to any development in treatment is a better understanding of the mechanisms of tissue injury underpinning progression which will in turn allow the identification of new targets against which treatments can be directed. There are additional complications in determining when progression actually starts, determining the impact of aging and defining the progressive clinical phenotypes – an area which has become increasingly complex in recent months. Evaluating potential new treatments in progressive MS also poses particular challenges including trial design and the selection of appropriate clinical and imaging outcomes - in particular, identifying an imaging biomarker for phase II trials of progressive MS. Despite these challenges, considerable progress is being made in developing new treatments targeting the innate immune system and exploring neuroprotective strategies. Further advances are being driven by a number of international networks, funded by the Progressive MS Alliance. Overall we are seeing encouraging progress as a result of co-ordinated global collaboration which offers real possibilities for truly effective treatment of progression.
MR Biomarkers in Spinocerebellar Ataxias
Magnetic Resonance Measures of Brain Blood Vessels, Metabolic Activity, and Pathology in Multiple Sclerosis
The normally functioning blood-brain barrier (BBB) regulates the transfer of material between blood and brain. BBB dysfunction has long been recognized in multiple sclerosis (MS), and there is considerable interest in quantifying functional aspects of brain blood vessels and their role in disease progression. Parenchymal water content and its association with volume regulation is important for proper brain function, and is one of the key roles of the BBB. There is convincing evidence that the astrocyte is critical in establishing and maintaining a functional BBB and providing metabolic support to neurons. Increasing evidence suggests that functional interactions between endothelia, pericytes, astrocytes, and neurons, collectively known as the neurovascular unit, contribute to brain water regulation, capillary blood volume and flow, BBB permeability, and are responsive to metabolic demands. Increasing evidence suggests altered metabolism in MS brain which may contribute to reduced neuro-repair and increased neurodegeneration. Metabolically relevant biomarkers may provide sensitive readouts of brain tissue at risk of degeneration, and magnetic resonance offers substantial promise in this regard. Dynamic contrast enhanced MRI combined with appropriate pharmacokinetic modeling allows quantification of distinct features of BBB including permeabilities to contrast agent and water, with rate constants that differ by six orders of magnitude. Mapping of these rate constants provides unique biological aspects of brain vasculature relevant to MS.
Associations between brain interoceptive network dysconnectivity and heightened peripheral inflammation in depression
Are the immune system, brain, mind and mood related? Could this explain why chronic low-grade peripheral inflammation is also noted in approximately 1/3 of those with major depressive disorder (MDD)? The field recognized today as immunopsychiatry was founded on scientific evidence that germinated over 30 years ago. Since, it has been understood that (i) there could be a causal link between inflammation and depression, (ii) select blood immune markers show robust potential as biomarkers for inflammation-linked depression, and more generally, (iii) Descartes' theories on mind-body dualism were biologically erroneous. Nonetheless, the mechanistic brain-immune axis in the trinity formulating inflammation-linked depression i.e. psycho-neuro-immunology, still remains unclear. This talk will discuss findings from our recent investigation endeavored to unpack this by linking functional connectivity abnormalities with peripheral immune markers.
Towards better interoceptive biomarkers in computational psychiatry
Empirical evidence and theoretical models both increasingly emphasize the importance of interoceptive processing in mental health. Indeed, many mood and psychiatric disorders involve disturbed feelings and/or beliefs about the visceral body. However, current methods to measure interoceptive ability are limited in a number of ways, restricting the utility and interpretation of interoceptive biomarkers in psychiatry. I will present some newly developed measures and models which aim to improve our understanding of disordered brain-body interaction in psychiatric illnesses.
Progressive Supranuclear Palsy – Update on Diagnostics, Biomarkers and Therapies
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.
Biomarkers for Addiction Treatment Development: fMRI Drug Cue Reactivity as an Example
This webinar is mainly focused on “Biomarkers for Addiction Treatment Development: fMRI Drug Cue Reactivity as an Example”. Biomarkers and Biotypes of Drug Addiction: funding opportunities at NIDA, Tanya Ramey (NIDA, US) Neuroimaging-based Biomarker Development for Clinical Trials, Owen Carmicheal (Pennington Biomedical Research Center, USA) ENIGMA-Addiction Cue Reactivity Initiative (ACRI) and Checklist, Hamed Ekhtiari (Laureate Institute for Brain Research, USA) ENIGMA-ACRI Checklist: Participant Characteristics, General fMRI Information, General Task Information, Cue Information, Task-related Assessments, Pre-Post Scanning Consideration (James Prisciandaro, Medical University of South Carolina, USA; Marc Kaufman, McLean Hospital/Harvard Medical School, USA; Anna Zilverstand, University of Minnesota; Torsten Wüstenberg, Charité Medical University Berlin, Germany; Falk Kiefer, University of Heidelberg, Germany; Amy Janes, Harvard Medical School, USA) How to Add fMRI Drug Cue Reactivity to the ENIGMA Consortium: Road Ahead, Hugh Garavan, University of Vermont)
NeuroCOVID: Epidemiology, biomarkers, and pathophysiology
Emergent scientists discuss Alzheimer's disease
This seminar is part of our “Emergent Scientists” series, an initiative that provides a platform for scientists at the critical PhD/postdoc transition period to share their work with a broad audience and network. Summary: These talks cover Alzheimer’s disease (AD) research in both mice and humans. Christiana will discuss in particular the translational aspects of applying mouse work to humans and the importance of timing in disease pathology and intervention (e.g. timing between AD biomarkers vs. symptom onset, timing of therapy, etc.). Siddharth will discuss a rare variant of Alzheimer’s disease called “Logopenic Progressive Aphasia”, which presents with temporo-parietal atrophy yet relative sparing of hippocampal circuitry. Siddharth will discuss how, despite the unusual anatomical basis underlying this AD variant, degeneration of the angular gyrus in the left inferior parietal lobule contributes to memory deficits similar to those of typical amnesic Alzheimer’s disease. Christiana’s abstract: Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder that causes severe deterioration of memory, cognition, behavior, and the ability to perform daily activities. The disease is characterized by the accumulation of two proteins in fibrillar form; Amyloid-β forms fibrils that accumulate as extracellular plaques while tau fibrils form intracellular tangles. Here we aim to translate findings from a commonly used AD mouse model to AD patients. Here we initiate and chronically inhibit neuropathology in lateral entorhinal cortex (LEC) layer two neurons in an AD mouse model. This is achieved by over-expressing P301L tau virally and chronically activating hM4Di DREADDs intracranially using the ligand dechloroclozapine. Biomarkers in cerebrospinal fluid (CSF) is measured longitudinally in the model using microdialysis, and we use this same system to intracranially administer drugs aimed at halting AD-related neuropathology. The models are additionally tested in a novel contextual memory task. Preliminary findings indicate that viral injections of P301L tau into LEC layer two reveal direct projections between this region and the outer molecular layer of dentate gyrus and the rest of hippocampus. Additionally, phosphorylated tau co-localize with ‘starter cells’ and appear to spread from the injection site. Preliminary microdialysis results suggest that the concentrations of CSF amyloid-β and tau proteins mirror changes observed along the disease cascade in patients. The disease-modifying drugs appear to halt neuropathological development in this preclincial model. These findings will lead to a novel platform for translational AD research, linking the extensive research done in rodents to clinical applications. Siddharth’s abstract: A distributed brain network supports our ability to remember past events. The parietal cortex is a critical member of this network, yet, its exact contributions to episodic remembering remain unclear. Neurodegenerative syndromes affecting the posterior neocortex offer a unique opportunity to understand the importance and role of parietal regions to episodic memory. In this talk, I introduce and explore the rare neurodegenerative syndrome of Logopenic Progressive Aphasia (LPA), an aphasic variant of Alzheimer’s disease presenting with early, left-lateralized temporo-parietal atrophy, amidst relatively spared hippocampal integrity. I then discuss two key studies from my recent Ph.D. work showcasing pervasive episodic and autobiographical memory dysfunction in LPA, to a level comparable to typical, amnesic Alzheimer’s disease. Using multimodal neuroimaging, I demonstrate how degeneration of the angular gyrus in the left inferior parietal lobule, and its structural connections to the hippocampus, contribute to amnesic profiles in this syndrome. I finally evaluate these findings in the context of memory profiles in other posterior cortical neurodegenerative syndromes as well as recent theoretical models underscoring the importance of the parietal cortex in the integration and representation of episodic contextual information.
Sparks, flames, and inferno: epileptogenesis in the glioblastoma microenvironment
Glioblastoma cells trigger pharmacoresistant seizures that may promote tumor growth and diminish the quality of remaining life. To define the relationship between growth of glial tumors and their neuronal microenvironment, and to identify genomic biomarkers and mechanisms that may point to better prognosis and treatment of drug resistant epilepsy in brain cancer, we are analyzing a new generation of genetically defined CRISPR/in utero electroporation inborn glioblastoma (GBM) tumor models engineered in mice. The molecular pathophysiology of glioblastoma cells and surrounding neurons and untransformed astrocytes are compared at serial stages of tumor development. Initial studies reveal that epileptiform EEG spiking is a very early and reliable preclinical signature of GBM expansion in these mice, followed by rapidly progressive seizures and death within weeks. FACS-sorted transcriptomic analysis of cortical astrocytes reveals the expansion of a subgroup enriched in pro-synaptogenic genes that may drive hyperexcitability, a novel mechanism of epileptogenesis. Using a prototypical GBM IUE model, we systematically define and correlate the earliest appearance of cortical hyperexcitability with progressive cortical tumor cell invasion, including spontaneous episodes of spreading cortical depolarization, innate inflammation, and xCT upregulation in the peritumoral microenvironment. Blocking this glutamate exporter reduces seizure load. We show that the host genome contributes to seizure risk by generating tumors in a monogenic deletion strain (MapT/tau -/-) that raises cortical seizure threshold. We also show that the tumor variant profile determines epilepsy risk. Our genetic dissection approach sets the stage to broadly explore the developmental biology of personalized tumor/host interactions in mice engineered with novel human tumor mutations in specified glial cell lineages.
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
Circadian/Multidien Molecular Oscillations and Rhythmicity of Epilepsy
The occurrence of seizures at specific times of the day has been consistently observed for centuries in individuals with epilepsy. Electrophysiological recordings provide evidence that seizures have a higher probability of occurring at a given time during the night and day cycle in individuals with epilepsy – the seizure rush hour. Which mechanisms underly such circadian rhythmicity of seizures? Why don’t they occur every day at the same time? Which mechanisms may underly their occurrence outside the rush hour? I shall present a hypothesis: MORE - Molecular Oscillations and Rhythmicity of Epilepsy, a conceptual framework to study and understand the mechanisms underlying the circadian rhythmicity of seizures and their probabilistic nature. The core of the hypothesis is the existence of circa 24h oscillations of gene and protein expression throughout the body in different cells and organs. The orchestrated molecular oscillations control the rhythmicity of numerous body events, such as feeding and sleep. The concept developed here is that molecular oscillations may favor seizure genesis at preferred times, generating the condition for a seizure rush hour. However, the condition is not sufficient, as other factors are necessary for a seizure to occur. Studying these molecular oscillations may help us understand seizure genesis mechanisms and find new therapeutic targets and predictive biomarkers. The MORE hypothesis can be generalized to comorbidities and the slower multidien (week/month period) rhythmicity of seizures.
Blood Biomarkers for Neurodegenerative Dementias: An Update
Autism spectrum disorder: from gene discovery to functional insights
Autism spectrum disorder (ASD) is a neurodevelopmental disorder affecting up to 1% of the population. Over the past few years, large-scale genomic studies have identified hundreds of genetic loci associated with liability to ASD. It is now time to translate these genetic discoveries into functional studies that can help us understand convergences and divergences across risk genes, and build pre-clinical cell and animal models. In this seminar, I will discuss some of the most recent findings on the genetic risk architecture of ASD. I will then expand on our work on biomarkers discovery and neurodevelopmental analyses in two rare genetic conditions associated with ASD: ADNP and DDX3X syndrome.
Neuroscience Investigations in the Virgin Lands of African Biodiversity
Africa is blessed with a rich diversity and abundance in rodent and avian populations. This natural endowment on the continent portends research opportunities to study unique anatomical profiles and investigate animal models that may confer better neural architecture to study neurodegenerative diseases, adult neurogenesis, stroke and stem cell therapies. To this end, African researchers are beginning to pay closer attention to some of her indigenous rodents and birds in an attempt to develop spontaneous laboratory models for homegrown neuroscience-based research. For this presentation, I will be showing studies in our lab, involving cellular neuroanatomy of two rodents, the African giant rat (AGR) and Greater cane rat (GCR), Eidolon Bats (EB) and also the Striped Owl (SO). Using histological stains (Cresyl violet and Rapid Golgi) and immunohistochemical biomarkers (GFAP, NeuN, CNPase, Iba-1, Collagen 2, Doublecortin, Ki67, Calbindin, etc), and Electron Microscopy, morphology and functional organizations of neuronal and glial populations of the AGR , GCR, EB and SO brains have been described, with our work ongoing. In addition, the developmental profiles of the prenatal GCR brains have been chronicled across its entire gestational period. Brains of embryos/foetuses were harvested for gross morphological descriptions and then processed using immunofluorescence biomarkers to determine the pattern, onset, duration and peak of neurogenesis (Pax6, Tbr1, Tbr2, NF, HuCD, MAP2) and the onset and peak of glial cell expressions and myelination in the prenatal GCR. The outcome of these research efforts has shown unique neuroanatomical expressions and networks amongst Africa’s rich biodiversity. It is hopeful that continuous effort in this regard will provide sufficient basic research data on neural developments and cellular neuroanatomy with subsequent translational consequences.
Oscillatory and fractal biomarkers of human memory
COSYNE 2022
Oscillatory and fractal biomarkers of human memory
COSYNE 2022
Blood biomarkers to monitor neuroinflammation: Insights from hematopoietic stem cell transplantation and gene therapy in X-linked adrenoleukodystrophy
FENS Forum 2024
Circulating microRNAs as biomarkers for Alzheimer’s disease
FENS Forum 2024
Circulating microRNAs and isomiRs as biomarkers for the initial insult and epileptogenesis in four experimental epilepsy models – The EPITARGET study
FENS Forum 2024
Computational analysis of Alzheimer’s disease-associated missense SNPs to understand underlying molecular mechanisms and identify diagnostic biomarkers
FENS Forum 2024
Early microstructural alterations in Alzheimer’s disease: novel MRI biomarkers of Aß deposition and glial activation in APP/PS1 mouse model
FENS Forum 2024
Effects of 5-HT2AR-mGluR2-based interventions on electrophysiological biomarkers in a rat model of alcohol addiction
FENS Forum 2024
Estimation of EEG gamma oscillation biomarkers: Exploring open thermodynamic cortical neurodynamics via the Rankine to Carnot cycles in healthy individuals and bipolar patients
FENS Forum 2024
Exploratory molecular biomarkers in adult spinal muscular atrophy patients treated with nusinersen
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 blood and neuronal biomarkers for disease onset and medication response in first-episode psychosis patients
FENS Forum 2024
Irisin levels in cerebrospinal fluid correlate with biomarkers and clinical dementia scores in Alzheimer’s disease
FENS Forum 2024
Eye movement abnormalities as early biomarkers of Alzheimer’s disease
FENS Forum 2024
Neuronal biomarkers associated with recovery of motor function in a mouse model of stroke
FENS Forum 2024
Novel potential biomarkers for multiple sclerosis: Evaluating the expression levels of miR-141, miR-9, MEG3, IFNG-AS1 and their relationship to different LINC00513 polymorphisms
FENS Forum 2024
Ocular biomarkers for early diagnosis of Alzheimer’s disease in non-human primate model
FENS Forum 2024
Psychophysiological biomarkers to assess the effectiveness of surface EMG biofeedback as an alternative therapy to reduce chronic low back pain
FENS Forum 2024
SLC6A4 and TPH2 methylation as potential biomarkers to inform antidepressant treatment choices
FENS Forum 2024
TNAP and P2X7R: New plasma biomarkers for Alzheimer's disease
FENS Forum 2024