Therapeutics
therapeutics
Prof Richard Smith
The Smith lab is seeking team members to conduct exciting research in human neurodevelopment and models of neuronal activity in the prenatal brain. Interested applicants can expect to work in an environment that promotes autonomy and all the resources to develop and expand the several ongoing research projects of the lab. These include, but are not limited to, questions relating to human brain development, human disease modeling (using high throughput approaches), and therapeutics. Current NIH funded projects are examining ion flux and biophysical properties of developing cell types in the prenatal brain, specifically as is relates to childhood diseases. As a trainee you will have to opportunity gain expertise in several state-of the art approaches widely used to interrogate important aspects of neurodevelopment, including human stem cell cerebral organoid models, single cell sequencing (RNA/ATAC), high-content confocal microscopy/screening, ferret model of cortex development and hiPSC derived neuronal models (excitatory, dopamine, inhibitory). Additional physiology approaches include, 2-photon imaging, high-throughput electrophysiology, patch-clamp, and calcium/voltage imaging. Please visit our website for details about our research, www.rsmithlab.com
Pharmacological exploitation of neurotrophins and their receptors to develop novel therapeutic approaches against neurodegenerative diseases and brain trauma
Neurotrophins (NGF, BDNF, NT-3) are endogenous growth factors that exert neuroprotective effects by preventing neuronal death and promoting neurogenesis. They act by binding to their respective high-affinity, pro-survival receptors TrkA, TrkB or TrkC, as well as to p75NTR death receptor. While these molecules have been shown to significantly slow or prevent neurodegeneration, their reduced bioavailability and inability to penetrate the blood-brain-barrier limit their use as potential therapeutics. To bypass these limitations, our research team has developed and patented small-sized, lipophilic compounds which selectively resemble neurotrophins’ effects, presenting preferable pharmacological properties and promoting neuroprotection and repair against neurodegeneration. In addition, the combination of these molecules with 3D cultured human neuronal cells, and their targeted delivery in the brain ventricles through soft robotic systems, could offer novel therapeutic approaches against neurodegenerative diseases and brain trauma.
The Promise of MitoQuinone Therapeutics in Experimental TBI
Brain-on-a-Chip: Advanced In Vitro Platforms for Drug Screening and Disease Modeling
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.
Molecular Mechanisms of Opioid Receptor-dependent Signaling and Novel Therapeutics,
Neurovascular Interactions: Mechanisms, Imaging, Therapeutics
Causal Symptom Network Mapping Based on Lesions and Brain Stimulation; Converging Evidence about a Depression Circuit Using Causal Sources of Information
It’s our pleasure to announce that we will host Shan Siddiqi and Michael D. Fox on Thursday, March 30th at noon ET / 6PM CET. Shan Siddiqi, MD, is an Assistant Professor of Psychiatry at Harvard Medical School and the director of Psychiatric Neuromodulation Research at the Brigham and Women’s Hospital. Michael D. Fox, MD, PhD, is an Associate Professor of Neurology at Harvard Medical School and the founding director of the Center for Brain Circuit Therapeutics at the Brigham and Women’s Hospital. The talks will be followed by a shared discussion. You can register via talks.stimulatingbrains.org to receive the (free) Zoom link!
Driving human visual cortex, visually and electrically
The development of circuit-based therapeutics to treat neurological and neuropsychiatric diseases require detailed localization and understanding of electrophysiological signals in the human brain. Electrodes can record and stimulate circuits in many ways, and we often rely on non-invasive imaging methods to predict the location to implant electrodes. However, electrophysiological and imaging signals measure the underlying tissue in a fundamentally different manner. To integrate multimodal data and benefit from these complementary measurements, I will describe an approach that considers how different measurements integrate signals across the underlying tissue. I will show how this approach helps relate fMRI and intracranial EEG measurements and provides new insights into how electrical stimulation influences human brain networks.
Redox and mitochondrial dysregulation in epilepsy
Epileptic seizures render the brain uniquely dependent on energy producing pathways. Studies in our laboratory have been focused on the role of redox processes and mitochondria in the context of abnormal neuronal excitability associated with epilepsy. We have shown that that status epilepticus (SE) alters mitochondrial and cellular redox status, energetics and function and conversely, that reactive oxygen species and resultant dysfunction can lead to chronic epilepsy. Oxidative stress and neuroinflammatory pathways have considerable crosstalk and targeting redox processes has recently been shown to control neuroinflammation and excitability. Understanding the role of metabolic and redox processes can enable the development of novel therapeutics to control epilepsy and/or its comorbidities.
Angelman syndrome: biomarkers and treatment opportunities
Elucidating the mechanism underlying Stress and Caffeine-induced motor dysfunction using a mouse model of Episodic Ataxia Type 2
Episodic Ataxia type 2 (EA2), caused by mutations in the CACNA1A gene, results in a loss-of-function of the P/Q type calcium channel, which leads to baseline ataxia, and attacks of dyskinesia, that can last a few hours to a few days. Attacks are brought on by consumption of caffeine, alcohol, and physical or emotional stress. Interestingly, caffeine and stress are common triggers among other episodic channelopathies, as well as causing tremor or shaking in otherwise healthy adults. The mechanism underlying stress and caffeine induced motor impairment remains poorly understood. Utilizing behavior, and in vivo and in vitro electrophysiology in the tottering mouse, a well characterized mouse model of EA2, or WT mice, we first sought to elucidate the mechanism underlying stress-induced motor impairment. We found stress induces attacks in EA2 though the activation of cerebellar alpha 1 adrenergic receptors by norepinephrine (NE) through casein kinase 2 (CK2) dependent phosphorylation. This decreases SK2 channel activity, causing increased Purkinje cell irregularity and motor impairment. Knocking down or blocking CK2 with an FDA approved drug CX-4945 prevented PC irregularity and stress-induced attacks. We next hypothesized caffeine, which has been shown to increase NE levels, could induce attacks through the same alpha 1 adrenergic mechanism in EA2. We found caffeine increases PC irregularity and induces attacks through the same CK2 pathway. Block of alpha 1 adrenergic receptors, however, failed to prevent caffeine-induced attacks. Caffeine instead induces attacks through the block of cerebellar A1 adenosine receptors. This increases the release of glutamate, which interacts with mGluR1 receptors on PC, resulting in erratic firing and motor attacks. Finally, we show a novel direct interaction between mGluR1 and CK2, and inhibition of mGluR1 prior to initiation of attack, prevents the caffeine-induced increase in phosphorylation. These data elucidate the mechanism underlying stress and caffeine-induced motor impairment. Furthermore, given the success of CX-4945 to prevent stress and caffeine induced attacks, it establishes ground-work for the development of therapeutics for the treatment of caffeine and stress induced attacks in EA2 patients and possibly other episodic channelopathies.
Psychedelics and related plasticity-promoting neurotherapeutics
Dr. David E. Olson will give a talk addressed to the Humanitas University Undergraduate Neurological Society students, focusing on his work on psychedelic drugs and related plasticity-promoting neurotherapeutics. The event will begin with a general and brief introduction to the topic by the HUUNS members.
Network science and network medicine: New strategies for understanding and treating the biological basis of mental ill-health
The last twenty years have witnessed extraordinarily rapid progress in basic neuroscience, including breakthrough technologies such as optogenetics, and the collection of unprecedented amounts of neuroimaging, genetic and other data relevant to neuroscience and mental health. However, the translation of this progress into improved understanding of brain function and dysfunction has been comparatively slow. As a result, the development of therapeutics for mental health has stagnated too. One central challenge has been to extract meaning from these large, complex, multivariate datasets, which requires a shift towards systems-level mathematical and computational approaches. A second challenge has been reconciling different scales of investigation, from genes and molecules to cells, circuits, tissue, whole-brain, and ultimately behaviour. In this talk I will describe several strands of work using mathematical, statistical, and bioinformatic methods to bridge these gaps. Topics will include: using artificial neural networks to link the organization of large-scale brain connectivity to cognitive function; using multivariate statistical methods to link disease-related changes in brain networks to the underlying biological processes; and using network-based approaches to move from genetic insights towards drug discovey. Finally, I will discuss how simple organisms such as C. elegans can serve to inspire, test, and validate new methods and insights in networks neuroscience.
JAK/STAT regulation of the transcriptomic response during epileptogenesis
Temporal lobe epilepsy (TLE) is a progressive disorder mediated by pathological changes in molecular cascades and neural circuit remodeling in the hippocampus resulting in increased susceptibility to spontaneous seizures and cognitive dysfunction. Targeting these cascades could prevent or reverse symptom progression and has the potential to provide viable disease-modifying treatments that could reduce the portion of TLE patients (>30%) not responsive to current medical therapies. Changes in GABA(A) receptor subunit expression have been implicated in the pathogenesis of TLE, and the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway has been shown to be a key regulator of these changes. The JAK/STAT pathway is known to be involved in inflammation and immunity, and to be critical for neuronal functions such as synaptic plasticity and synaptogenesis. Our laboratories have shown that a STAT3 inhibitor, WP1066, could greatly reduce the number of spontaneous recurrent seizures (SRS) in an animal model of pilocarpine-induced status epilepticus (SE). This suggests promise for JAK/STAT inhibitors as disease-modifying therapies, however, the potential adverse effects of systemic or global CNS pathway inhibition limits their use. Development of more targeted therapeutics will require a detailed understanding of JAK/STAT-induced epileptogenic responses in different cell types. To this end, we have developed a new transgenic line where dimer-dependent STAT3 signaling is functionally knocked out (fKO) by tamoxifen-induced Cre expression specifically in forebrain excitatory neurons (eNs) via the Calcium/Calmodulin Dependent Protein Kinase II alpha (CamK2a) promoter. Most recently, we have demonstrated that STAT3 KO in excitatory neurons (eNSTAT3fKO) markedly reduces the progression of epilepsy (SRS frequency) in the intrahippocampal kainate (IHKA) TLE model and protects mice from kainic acid (KA)-induced memory deficits as assessed by Contextual Fear Conditioning. Using data from bulk hippocampal tissue RNA-sequencing, we further discovered a transcriptomic signature for the IHKA model that contains a substantial number of genes, particularly in synaptic plasticity and inflammatory gene networks, that are down-regulated after KA-induced SE in wild-type but not eNSTAT3fKO mice. Finally, we will review data from other models of brain injury that lead to epilepsy, such as TBI, that implicate activation of the JAK/STAT pathway that may contribute to epilepsy development.
Improving Communication With the Brain Through Electrode Technologies
Over the past 30 years bionic devices such as cochlear implants and pacemakers, have used a small number of metal electrodes to restore function and monitor activity in patients following disease or injury of excitable tissues. Growing interest in neurotechnologies, facilitated by ventures such as BrainGate, Neuralink and the European Human Brain Project, has increased public awareness of electrotherapeutics and led to both new applications for bioelectronics and a growing demand for less invasive devices with improved performance. Coupled with the rapid miniaturisation of electronic chips, bionic devices are now being developed to diagnose and treat a wide variety of neural and muscular disorders. Of particular interest is the area of high resolution devices that require smaller, more densely packed electrodes. Due to poor integration and communication with body tissue, conventional metallic electrodes cannot meet these size and spatial requirements. We have developed a range of polymer based electronic materials including conductive hydrogels (CHs), conductive elastomers (CEs) and living electrodes (LEs). These technologies provide synergy between low impedance charge transfer, reduced stiffness and an ability to be provide a biologically active interface. A range of electrode approaches are presented spanning wearables, implantables and drug delivery devices. This talk outlines the materials development and characterisation of both in vitro properties and translational in vivo performance. The challenges for translation and commercial uptake of novel technologies will also be discussed.
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.
Epigenetic regulation of alternative splicing in the context of cocaine reward
Neuronal alternative splicing is a key gene regulatory mechanism in the brain. However, the spliceosome machinery is insufficient to fully specify splicing complexity. In considering the role of the epigenome in activity-dependent alternative splicing, we and others find the histone modification H3K36me3 to be a putative splicing regulator. In this study, we found that mouse cocaine self-administration caused widespread differential alternative splicing, concomitant with the enrichment of H3K36me3 at differentially spliced junctions. Importantly, only targeted epigenetic editing can distinguish between a direct role of H3K36me3 in splicing and an indirect role via regulation of splice factor expression elsewhere on the genome. We targeted Srsf11, which was both alternatively spliced and H3K36me3 enriched in the brain following cocaine self-administration. Epigenetic editing of H3K36me3 at Srsf11 was sufficient to drive its alternative splicing and enhanced cocaine self-administration, establishing the direct causal relevance of H3K36me3 to alternative splicing of Srsf11 and to reward behavior.
Pitt-Hopkins Syndrome, mouse models, neurodevelopment, therapeutics
Human stem cell models of neurodegeneration: complex, relevant and robust
Microbiome and behaviour: Exploring underlying mechanisms
Environmental insults alter brain function and behaviour inboth rodents and people. One putative underlying mechanism that has receivedsubstantial attention recently is the gut microbiota, the ecosystem ofsymbiotic microorganisms that populate the intestinal tract, which is known toplay a role in brain health and function via the gut-brain axis. Two keyenvironmental insults known to affect both brain function and behaviour, andthe gut microbiome, are poor diet and psychological stress. While there isstrong evidence for interactions between the microbiome and host physiology inthe context of chronic stress, little is known about the role of the microbiomein the host response to acute stress. Determining the underlying mechanisms bywhich stress may provoke functional changes in the gut and brain is criticalfor developing therapeutics to alleviate adverse consequences of traumaticstress.
Regenerative Neuroimmunology - a stem cell perspective
There are currently no approved therapies to slow down the accumulation of neurological disability that occurs independently of relapses in multiple sclerosis (MS). International agencies are engaging to expedite the development of novel strategies capable of modifying disease progression, abrogating persistent CNS inflammation, and support degenerating axons in people with progressive MS. Understanding why regeneration fails in the progressive MS brain and developing new regenerative approaches is a key priority for the Pluchino Lab. In particular, we aim to elucidate how the immune system, in particular its cells called myeloid cells, affects brain structure and function under normal healthy conditions and in disease. Our objective is to find how myeloid cells communicate with the central nervous system and affect tissue healing and functional recovery by stimulating mechanisms of brain plasticity mechanisms such as the generation of new nerve cells and the reduction of scar formation. Applying combination of state-of-the-art omic technologies, and molecular approaches to study murine and human disease models of inflammation and neurodegeneration, we aim to develop experimental molecular medicines, including those with stem cells and gene therapy vectors, which slow down the accumulation of irreversible disabilities and improve functional recovery after progressive multiple sclerosis, stroke and traumatic injuries. By understanding the mechanisms of intercellular (neuro-immune) signalling, diseases of the brain and spinal cord may be treated more effectively, and significant neuroprotection may be achieved with new tailored molecular therapeutics.
Understanding the cellular and molecular landscape of autism spectrum disorders
Large genomic studies of individuals with autism spectrum disorders (ASD) have revealed approximately 100-200 high risk genes. However, whether these genes function in similar or different signaling networks in brain cells (neurons) remains poorly studied. We are using proteomic technology to build an ASD-associated signaling network map as a resource for the Autism research community. This resource can be used to study Autism risk genes and understand how pathways are convergent, and how patient mutations change the interaction profile. In this presentation, we will present how we developed a pipeline using neurons to build protein-protein interaction profiles. We detected previously unknown interactions between different ASD risk genes that have never been linked together before, and for some genes, we identified new signaling pathways that have not been previously reported. This resource will be available to the research community and will foster collaborations between ASD researchers to help accelerate therapeutics for ASD and related disorders.
Gene Therapy for Neurodegeneration
One of the major challenges in developing therapeutics for the neurodegenerative disorders is the blood-brain barrier, limiting the availability of systemically administered therapies such as recombinant proteins or monoclonal antibodies from reaching the brain. Direct central nervous system (CNS) gene therapy using adeno-associated virus vectors expressing a therapeutic protein, monoclonal antibody or inhibiting RNA-coding sequences has two characteristics ideal for therapy of neurodegenerative disorders: circumventing the blood-brain barrier by directly expressing the therapy in the brain and the ability to provide persistent therapy with only a single administration. There are several critical parameters relevant to successful CNS gene therapy, including choice of vector, design of the gene to be expressed, delivery/route of administration, dose and anti-vector immune responses. The presentation will focus on these issues, the current status of clinical trials of gene therapy for neurodegeneration and specific challenges that will need to be overcome to ensure the success of these therapies.
Towards therapeutics for Autism Spectrum Disorder using Syngap1 heterozygous mouse model
Targeting the Endocannabinoid System for Management of Chemotherapy, HIV and Antiretroviral-Induced Neuropathic Pain
Chemotherapeutic drugs (used for treating cancer), HIV infection and antiretroviral therapy (ART) can independently cause difficult-to-manage painful neuropathy. Paclitaxel, a chemotherapeutic drug, for example is associated with high incidence of peripheral neuropathy, around 71% of the patients of which 27% of these develop neuropathic pain. Use of cannabis or phytocannabinoids has been reported to improve pain measures in patients with neuropathic pain, including painful HIV-associated sensory neuropathy and cancer pain. Phytocannabinoids and endocannabinoids, such as anandamide and 2-arachidonoylglycerol (2-AG), produce their effects via cannabinoid (CB) receptors, which are present both in the periphery and central nervous system. Endocannabinoids are synthesized in an “on demand” fashion and are degraded by various enzymes such as fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MGL). Various studies, including those from our group, suggest that there are changes in gene and protein expression of endocannabinoid molecules during chemotherapy-induced neuropathic pain (CINP), HIV and antiretroviral-induced neuropathic pain. Analysis of endocannabinoid molecule expression in the brain, spinal cord and paw skin using LC-MS/MS show that there is a specific deficiency of the endocannabinoids 2-AG and/or anandamide in the periphery during CINP. Various drugs including endocannabinoids, cannabidiol, inhibitors of FAAH and MGL, CB receptor agonists, desipramine and coadministered indomethacin plus minocycline have been found to either prevent the development and/or attenuate established CINP, HIV and antiretroviral-induced neuropathic pain in a CB receptor-dependent manner. The results available suggest that targeting the endocannabinoid system for prevention and treatment of CINP, HIV-associated neuropathic pain and antiretroviral-induced neuropathic pain is a plausible therapeutic option.
Vagal sensory neurons that guard the airways
The vagus nerve contains a diversity of sensory neurons that detect peripheral stimuli such as blood pressure changes at the aortic arch, lung expansion during breathing, meal-induced stomach distension, and chemotherapeutics that induce nausea. Underlying vagal sensory mechanisms are largely unresolved at a molecular level, presenting tremendously important problems in sensory biology. We charted vagal sensory neurons by single cell RNA sequencing, identifying novel cell surface receptors and classifying a staggering diversity of sensory neuron types. We then generated a collection of ires-Cre knock-in mice to target each neuron type, and adapted genetic tools for Cre-based anatomical mapping, in vivo imaging, targeted ablation, and optogenetic control of vagal neuron activity. We found different sensory neuron types that innervate the lung and exert powerful effects on breathing, others that monitor and control the digestive system, and yet others that innervate that innervate the larynx and protect the airways. Together with Ardem Patapoutian, we also identified a critical role for Piezo mechanoreceptors in the sensation of airway stretch, which underlies a classical respiratory reflex termed the Hering-Breuer inspiratory reflex, as well as in the neuronal sensation of blood pressure and the baroreceptor reflex.