Skip to Main Content

2020-2023 Project Descriptions

Cell-type-specific Proteomics in Human Brains Using Immunopanning and Antibody-Based Proximity Labeling

Yifei Cai, Department of Neurology, Yale University

In the realm of neuroscience, the quest for comprehending the intricacies of the brain at a cell-type- specific and single-cell resolution is steadfast. However, this pursuit is impeded by the challenges posed by achieving accurate tissue isolation and acquiring fresh brain samples. Thus far, cell-type-specific human brain proteomes remain elusive. This poses a substantial knowledge gap in understanding protein expression and RNA-protein correlation at the cell-type-specific and single-cell levels. Moreover, the absence of reference proteomes tailored to distinct cell types within the human brain accentuates this knowledge gap, as these reference proteomic data are essential for emerging fields like subcellular proteomics and single-cell proteomics. In this pilot project, I will establish two parallel isolation and proteome methods to study cell-type-specific proteomics in both fresh and postmortem human brain samples. Firstly, I will employ innovative immunopanning techniques to isolate different cell types in fresh human brains. Subsequently, cell-type specific DDA- and DIA-based LC-MS-MS proteomics will be applied to analyze these samples. To address potential protein loss during tissue isolation, I will utilize a novel antibody-based proximity labeling proteomics pipeline that I previously established to uncover proteins in cell-type-specific compartments, such as cell bodies and processes, in postmortem human brain sections. By comparing the proteome datasets obtained from immunopanning and proximity labeling in fresh and postmortem brain samples, I aim to gain insights into the cell-type-specific proteome in human brains. Ultimately, this work seeks to establish high-quality cell-type-specific reference proteomes in human brains. These reference datasets will be instrumental in understanding the molecular basis of human brains, illuminating RNA-protein correlations, and advancing data analysis in subcellular proteomics and single-cell proteomics in humans. Furthermore, the project will provide valuable tools and resources to enhance our understanding of human brains in both health and diseases, such as drug addiction.

Investigating the Cell Type-Specific Recruitment of the SAGA Complex in Cocaine Use Disorder

Soren Emerson, Department of Pharmacology, Vanderbilt U.

Cocaine use disorder (CUD) imposes a large burden on public health, particularly because there are no FDA-approved pharmacotherapies for the disorder. The onset and maintenance of CUD is driven by physiological and molecular changes within the brain that lead to maladaptive behavior associated with cocaine-taking and cocaine-seeking. A key neuronal population in this dysregulation is dopamine 1 receptor expressing medium spiny neurons (D1 MSNs) in the nucleus accumbens (NAc). These cells are activated by acute cocaine, undergo physiological and transcriptional plasticity following repeated cocaine exposure, and are recruited by cocaine-associated cues to drive drug-seeking and self-administration. Although a causal role in drug-induced behavior has been identified, the molecular mechanisms underlying cocaine-induced transcription dysregulation remain poorly understood. Lysine acetyltransferase 2a (KAT2a) is a critical cocaine-induced epigenetic regulator in the NAc, and the Spt-Ada-Gcn5-Acetyltransferase (SAGA) complex is a regulator of stimulus-responsive and cell type-specific gene expression. The goal of this proposal is to characterize KAT2a recruitment and assembly within the SAGA complex in NAc D1 MSNs following cocaine self-administration. We will test the hypothesis that cocaine self-administration recruits KAT2a assembly within the SAGA complex in NAc D1 MSNs.

We present in our preliminary data a detailed series of proteomic and bioinformatic studies through which we identified KAT2a–the acetyltransferase subunit of the SAGA complex–as an upstream regulator of the wide-scale transcriptional dysregulation associated with cocaine exposure in the NAc of both males and females. We also show that a mutation of KAT2a that impairs its function only in D1 MSNs greatly impairs cocaine self-administration. We hypothesize that KAT2a acts as a component of the SAGA complex within NAc D1 MSNs to control the transcriptional plasticity that drives cocaine self-administration. To address this question, we will collaborate with the Yale/NIDA Neuroproteomics Center to conduct Rapid Immunoprecipitation Mass Spectrometry of Endogenous Proteins (RIME) to assess SAGA complex formation following cocaine self-administration in genetic mouse lines that allow for cell-type specific isolation of the SAGA complex from NAc D1 MSNs. The overall goal is to characterize cocaine-induced KAT2a recruitment and SAGA complex formation selectivity within NAc D1 MSNs. In Aim 1, we will express wildtype KAT2a fused to a non-endogenously expressed V5 peptide tag selectively within NAc D1 MSNs and characterize the interacting partners of KAT2a in vivo and the effect that cocaine self-administration exerts on these protein-protein associations compared to saline control. In Aim 2, we will conditionally express a V5-tagged KAT2a mutant that prevents KAT2a recruitment to chromatin by cocaine and assess the effect of this mutation on cocaine-induced recruitment and formation of the SAGA complex. The experimental goals in this pilot grant will provide the technical capability necessary to characterize the proteomic mechanisms underlying the cell type-specific epigenetic and transcriptional mechanisms underlying substance use disorder In addition, the experimental findings will define a cell type-specific neuro epigenetic mechanism of CUD.

Sex-specific Proteomic Adaptations in Microglia in Response to Nicotine Treatment and Withdrawal

Nadine Kabbani, School of Systems Biology, George Mason University

Microglia, the resident immune cells of the brain, respond to environmental cues through morphological, transcriptional, and metabolic signaling adaptations. Throughout their life span microglia dynamically participate in many neural processes from synaptic development to functional and structural plasticity through complex patterns of neuroinflammatory signaling between microglia and other neural cells. Evidence indicates that microglia responses are important in drug-reward behaviors and contribute to addiction to various drugs including alcohol, cocaine, opioid, and nicotine. The conceptual premise of this project is that microglia are drivers of innate immune memory within critical neural circuits that mediate drug-associated behaviors. In support of this, there is evidence that although microglia are widely distributed throughout the adult brain, higher expression has been reported in reward centers such as the substantia nigra, basal ganglia, and hippocampus. In collaboration with ongoing research on sex-specific mechanisms of nicotine addiction and withdrawal at the Yale/NIDA Neuroproteomics Center, this project will identify proteomic changes in microglia of adult mice. Initial work will focus on the hippocampus as an important site for sex-specific neuroinflammatory signaling during nicotine addiction and withdrawal.

Understanding Biased CB1R Signaling Through Phosphoproteomics

Angela Henderson Redmond, Department of Biomedical Sciences, Marshall University

Of the 50 million+ adults in the United States suffering from chronic pain, 19.6 million suffer from high-impact chronic pain that interferes with life or work activities. Though cannabinoid-based therapies offer a needed alternative to opioids for the treatment of chronic pain, tolerance to their therapeutic effects can rapidly develop, limiting their efficacy while also facilitating escalating consumption and dependence. Consequently, identification of the mechanism(s) of tolerance to the analgesic effects of cannabinoids may serve to prolong their clinical utility. Desensitization (uncoupling of a receptor from its G proteins) and internalization (loss of receptor from the cell surface) are cell-based correlates of tolerance that involve G protein-coupled receptor kinase phosphorylation. For cannabinoid type 1 receptors (CB1R), desensitization and internalization appear to be mediated by distinct receptor domains. Previous work in vitro suggests that desensitization of CB1R is mediated by residues S426 and S430 while six different carboxy terminal serines and threonines (T461, S463, S465, T466, T468, and S469) are critical for CB1R internalization. We subsequently evaluated the contribution of these 8 different phosphorylation sites in vivo through the creation first of a desensitization-resistant (S426A/S430A) and then an internalization-resistant six-point mutant (6PM) mouse by mutating each of the phosphorylation sites to a non-phosphorylated alanine. The central hypothesis is that internalization of CB1R counteracts tolerance to strongly internalizing cannabinoids, such as CP55,940, and that tolerance to CP55,940 would be more profoundly impacted in 6PM mice relative to Δ9-THC, a weakly internalizing, partial agonist. In contrast, desensitization of CB1R would have a greater effect on tolerance development to Δ9-THC compared to CP55,940. Behaviorally, we have repeatedly shown that desensitization-resistant S426A/S430A mice display enhanced sensitivity and delayed tolerance to Δ9-THC-mediated antinociception. Preliminary data collected from our recently generated six-point mutants (T461A/S463A/S465A/T466A/T468A/S469A; 6PM) has revealed decreased sensitivity and faster tolerance to CP55,940-induced antinociception. This proposal seeks to identify molecular neuroadaptations associated with cannabinoid tolerance that will complement our behavioral data, to be utilized in a larger proposal for NIDA funding on cannabinoid tolerance. Specifically, this pilot project will utilize the cutting-edge use of phosphoproteomics to identify agonist-specific downstream targets in commercially available HEK293 cells stably expressing HA-CB1R that are treated with either vehicle, Δ9-THC or CP55,940 at time points (5 and 60 minutes) sufficient to induce desensitization and/or internalization. Our hypothesis is that treatment with CP55,940, a strongly internalizing cannabinoid agonist, will result in robust phosphorylation of the six internalization residues while treatment with Δ9-THC, a weakly internalizing agonist, will result in phosphorylation at the residues involved in desensitization, S426 and S430, with limited phosphorylation at the six internalization residues. The results of this project will be utilized in a larger grant to devise the precise nature of diverse downstream signaling pathways dictated through CB1R agonist-specific binding with the eventual aim of devising appropriate therapeutic strategies to combat cannabis tolerance and dependence in managing chronic pain.

Investigating the Central Amygdala Neuronal Proteome Mediating the Protective Effect of Social Reward on Incubation of Heroin Craving

Alexandra Fall, Department of Anatomy and Neurobiology, University of Maryland

Despite strides toward understanding circuit and molecular mechanisms of substance use disorders (SUDs), treatment options remain largely unchanged. This impasse is due, in part, to limitations in the construct and predictive validity of animal models of drug self-administration and relapse, which rarely incorporate social factors. In both humans and laboratory animals, adverse social interactions and social isolation promote drug self-administration and relapse, while positive social interaction tends to be protective. We recently developed an operant rat model of choice between drugs and social interaction and showed the profound protective effects of the latter on drug self-administration and relapse. Our research revealed two major findings. First, rats strongly prefer operant social interaction over heroin, methamphetamine (Meth), and cocaine. Additionally, social choice-induced abstinence (voluntary abstinence) decreases incubation (the progressive increase in drug seeking during abstinence) of heroin craving and prevents incubation of Meth craving. This protective effect was associated with activation of protein kinase-Cδ (PKCδ) in the central amygdala lateral part (CeL). In contrast, after forced abstinence, the reliable expression of incubation of Meth craving was associated with activation of CeL-somatostatin (SOM). The cellular and molecular mechanisms mediating the protective effect of social reward on heroin craving after voluntary abstinence remain unknown. Based on our preliminary data, we hypothesize that molecular changes within CeL PKCδ-expressing neurons in CeL are selective for the social-based buffering of incubation of heroin craving. In contrast, molecular changes within SOM-expressing neurons are selective for the forced abstinence-promoting expression of incubation of heroin craving. The overarching aim of this proposal is to elucidate changes in the neuronal proteome of CeL PKCδ/SOM- expressing neurons in heroin craving in order to understand the functional molecular pathways driving either the protective or promoting effects of voluntary versus forced abstinence procedures, respectively. By combining viral-mediated immunolabeling of CeL PKCδ-expressing neurons with laser capture microdissection (LCM) followed by isobaric tags for relative and absolute quantitation (iTRAQ), we predict differential protein expression as well as differential post translational modifications (PTMs) within the CeL PKCδ/SOM-expressing neurons during heroin craving after either voluntary or forced abstinence. Our proposal will provide new insights into the molecular mechanisms mediating the protective effect of social reward on addiction related measures merging cutting-edge tools for the investigation of neural circuits with an original behavioral model. This direction will provide collaborative potential and introduce new ideas and conceptual frameworks to the Yale/NIDA Neuroproteomics Center’s repertoire with potential direct translational applicability.

Proteomic Profiling of Glutamate Neurotransmission Alterations

with Chronic THC and/or Ethanol Exposure in the Context of Addiction in iPSC-Derived Human Excitatory Neurons

Isabel Gameiro-Ros, Department of Neuroscience, Icahn School of Medicine at Mount Sinai

Addiction is a chronic relapsing disease with devastating consequences to the health and well-being of patients and their families. Addiction is frequently not restricted to one drug: cannabis and alcohol are amongst the two most concomitantly used and abused drugs. Addiction vulnerability has been associated with altered prefrontal cortex (PFC) activity, leading to a more impulsive executive function. Repeated drug exposure increases glutamate release by PFC glutamatergic neurons projecting to NAc, increasing the sensitivity of the reward circuit to prior drug use-associated environmental cues. Previous in vivo studies identified important signaling networks involved in drug-seeking behaviors, but focused on VTA and NAc rather than PFC, and did not clarify whether the observed changes map directly onto changes in the protein machinery of the neurons. Moreover, addiction has a strong inheritable component, but animal models cannot reproduce the genetic environment of individuals with addiction or that are vulnerable to develop substance abuse. Using human iPSC-derived glutamatergic neurons as a model system of human cortical excitatory neurons is a suitable strategy to overcome the limitations of animal models.

Combining GWAS summary statistics and addiction related traits, our collaborators from the Collaborative Studies on Genetics of Alcoholism (COGA) have calculated polygenic risk scores (PRS) for a cohort of healthy individuals, as well as for cannabis and alcohol addicted individuals, generating an addiction-PRS distribution. hiPSCs from these individuals will be differentiated into excitatory neurons using the NGN2-induction protocol and chronically exposed to THC, ethanol, a combination of both drugs, or vehicle. The goal of the present study is to identify protein changes in the synaptic machinery of the neurons due to chronic THC and/or ethanol exposure using cutting-edge iTRAQ proteomics, and compare this changes between individuals with high and low addiction-PRS. The findings of this study will be further integrated with transcriptomic and functional datasets that have been generated in this cohort of addiction-PRS individuals, to converge on drug-induced disruptions in glutamatergic signaling in human hiPSC-derived excitatory neurons. Elucidating the whole synaptosomal proteome alterations due to chronic THC and/or ethanol exposure in these hiPSC-derived glutamatergic neurons and evaluating the potential differences in these drug-associated changes between individuals with high and low PRS for addiction will markedly advance our understanding of the molecular underpinnings behind genetic predisposition to addiction.

Targeted Discovery and Characterization of Opioid Use Disorder (OUD) Causal Genes Through Proteomic Analysis of Human Brain Regions

Vena Martinez, Department of Psychiatry, Yale University

Opioid overdoses continue to increase, despite newly available treatments and enhanced legal regulations. New therapeutic targets are urgently needed. Opioid use disorder (OUD) is twice as likely to develop in PTSD patients, and OUD patients with PTSD have a greater risk of increased OUD severity. This suggests predisposing risk factors exist for the development and severity of OUD in patients with PTSD and perhaps other psychiatric disorders. The amygdala is a significant brain region of convergence between OUD and PTSD, regulating positive and negative emotional states for both conditions. Additionally, altered amygdala structure and volume have been reported in both conditions. We previously conducted transcriptomic profiles in human postmortem amygdala and identified differentially expressed transcripts for both OUD and PTSD. In collaboration with Yale/NIDA Neuroproteomic Center’s Discovery Proteomics Core, we propose a pilot study to identify differentially expressed proteins and causal genes in OUD+, PTSD+, OUD+PTSD, and normal control (NC) postmortem amygdala. In Aim 1, we will generate comprehensive proteomic profiles for each condition. We will use our uniquely developed, state-of-the-art bioinformatic pipeline to integrate transcriptomic and proteomic profiles, and identify sex-specific differences. In addition, we will conduct proteomic co-expression analysis to identify cell type-specific differences in OUD. We will extend our studies to identify single cell type proteomic changes in Aim 2 by using laser capture microdissection on frozen sections of OUD and PTSD postmortem tissue. We will then conduct proteomic profiling and pathway analysis to identify dysregulated expression of neuronal and non-neuronal cells. These preliminary findings will identify biological factors that can be mechanistically interrogated in animal studies to potentially advance diagnostics and therapeutics of OUD.

Developing New Enzymatic Scaffolds for Proximity Labeling

Ken Loh, Department of Comparative Medicine, Yale University

Unbiased, proteomic approaches for defining the molecular components of a spatially defined region of interest often rely on long extensive purification schemes to isolate the region of interest. Proximity labeling based proteomic approaches have revolutionized this workflow for molecular discovery because they circumvent the need for classical forms of biochemical fractionation, allowing access to many previously unpurifiable subcellular structures. Most iterations of proximity labeling rely on the expression of an enzyme in the region of interest to generate a reactive intermediate that tags neighboring proteins with a biotin handle, for subsequent streptavidin enrichment and identification by proteomics. Although applied widely and successfully in vitro and in cell culture systems, proximity labeling suffers from several limitations when applied in vivo. The major limitation when applied in vivo arises from additional sources of background in tissue, particularly from endogenous biotinylated proteins that similarly compete for the same binding site during streptavidin enrichment. This limitation is particularly debilitating when the region of interest is a small subpopulation of cells within the tissue, such as a small subpopulation of neurons within the brain. This challenge could, however, be overcome by using non-biotin-streptavidin based approaches for chemical enrichment of labeled proteins. As a step towards expanding the in vivo applications of proximity labeling, this proposal will identify or engineer new enzyme scaffolds, capable of generating reactive intermediates like those generated by the proximity labeling enzyme TurboID, that have been functionalized for chemical enrichment instead of biotin-streptavidin enrichment.

Short- and Long-Term Proteomic Changes in Dendritic Spines Induced by Psychedelic Compounds with Therapeutic Applications

Sarah Jefferson, Department of Psychiatry, Yale University

Psychedelic drugs, which act on the serotonin-2a receptor, have recently generated interest as potential novel treatments for substance use and mood disorders. A single dose of psilocybin confers antidepressant effects lasting for several months in humans and rodent studies have demonstrated that psilocybin induces structural plasticity in the prefrontal cortex on a timescale which appears to correlate with its antidepressant effects. Causal manipulations of dendritic spine formation can occlude the rapidly acting antidepressant effect for other novel antidepressants like ketamine, suggesting its necessity. Despite great interest in this area, there continue to be fundamental questions about how these spines may integrate into circuits to produce therapeutic effects. Understanding how dendritic spines form and persist in response to rapidly acting antidepressants may facilitate the utilization of these compounds as therapeutics for substance use disorders and could lead to development of pharmacological strategies that prolong their therapeutic effects.

Interestingly, the duration of drug effects on plasticity may not correlate with duration of psychedelic effects. We have demonstrated that even a drug which elicits very brief psychedelic effects can lead to long-term structural plasticity. Using, in vivo two-photon dendritic spine imaging, we have shown that the psychedelic compound 5-methoxy-DMT (5-MeO-DMT) enhances spinogenesis for >1 month after a single dose. In contrast, prior studies have demonstrated that the rapidly acting antidepressant ketamine only enhances spinogenesis for up to 1 week, which correlates with the duration of antidepressant effects. The goal of this study is to investigate the synaptic proteome of newly generated and persistent spines after administration of psychedelics with differing temporal effects on plasticity. We propose to do so using a novel approach via proximity labeling of synaptic proteins in collaboration with Dr. Angus Nairn and the NIDA Proteomics Center. This pilot study will enable us to identify targets for future functional studies testing the necessity of these identified proteins in spinogenesis. Future studies will couple these proximity labeling approaches and findings with in vivo two photon imaging of neural structure and second messenger systems in order to identify spatiotemporal events in therapeutic spinogenesis.

Synaptoproteomic Correlates of Stress-induced Acetylcholine Release in mPFC: Mechanistic Evaluation of Depression/Addiction Co-morbidity

Zuhair I. Abdulla, Department of Psychiatry, Yale University

Major depression is highly comorbid with tobacco smoking, the leading cause of preventable death in the United States. Nicotinic acetylcholine receptors are a key subgroup of receptors involved in ACh signaling and are the primary target of nicotine, the addictive component of tobacco smoke. Importantly, increased ACh signaling is implicated in the etiology of depression and chronic nicotine use has been shown to desensitize nicotinic receptors, suggesting that cigarette used by people with depression is a form of self-medication. Increases in ACh are also important in learning, memory, and attentional processes, suggesting that optimal levels of ACh are beneficial while excessive increases are detrimental to affective health. In this scenario, excessively increased ACh during stressful events would lead to a negative encoding bias, in which stronger encoding occurs, leading to increased depressive symptoms and given the co-morbidity with smoking, increased tobacco use. To test this theory, I used the GRAB ACh4.3 sensor to record ACh transients in mice subjected to inescapable shocks as part of learned helplessness testing and found that mice classified as helpless in a later active avoidance test had increased ACh signaling in the medial prefrontal cortex relative to resilient mice. Furthermore, although the number of male and female mice classified as helpless was similar, the increased ACh signaling was more robust in male mice, a potentially important sex difference. Because synaptic dysfunctions in the mPFC are a hallmark of stress response and depression, I plan to use synaptosomal proteomics and phospho-proteomics to discover the differences between helpless and resilient mice responsible for alterations in ACh signaling. The findings from the study, along with previous work detailing the nicotinic ACh receptor-associated proteome will more clearly elucidate protein-dependent mechanisms contributing to the comorbidity of tobacco smoking and depression.

Mapping Methamphetamine-Induced Changes in the GIRK Channel Interaction Proteome

Xiaofan Li, Department of Neuroscience, Icahn School of Medicine at Mount Sinai

G protein-gated inwardly rectifying potassium (GIRK) channels are widely expressed in the brain and mediate GPCR-dependent slow inhibition. Malfunctioning and dysregulation of GIRK channels can result in various neurological and psychiatric disorders. Drugs of abuse can cause sustained alterations in GIRK function and trafficking in different brain regions, including the medial prefrontal cortex (mPFC), hippocampus and ventral tegmental area (VTA), important nodes in the brain reward pathway. In order to identify new therapeutic targets and develop more effective treatments for drug addiction, it is critical that we understand the molecular mechanisms behind such drug-induced plasticity. Our goal is to use the novel in vivo iBioID technique to define the GIRK channel interaction proteome (i.e., proteins in close proximity to GIRK channels in vivo) to discover novel regulators of GIRK channel function and trafficking. Furthermore, we will investigate how psychostimulant drugs such as methamphetamine alter the GIRK interaction proteome in a regional and cell type-specific manner. We expect to identify a few candidate proteins whose interaction with GIRK channels is modulated by repeated methamphetamine exposure, and we plan to investigate their functional significance with a new R01 grant in the future.

Proteomic Analysis of Protein Partners that Govern TRPA1 Trafficking and Functionality

Candice E. Paulsen, Department of Molecular Biophysics & Biochemistry, Yale University

Roughly one third of Americans suffer from chronic pain, yet little progress has been made to develop new pain remedies beyond non-steroidal anti-inflammatory agents and opioids. While opioids can be effective analgesic agents, these drugs harbor poor side effects and the risk of addiction because opioid receptors are expressed in neurons and brain regions outside the pain producing somatosensory neural network. An attractive therapeutic approach to develop new analgesic and anti-inflammatory agents, which will not harbor these poor side effects, is to identify receptors predominantly expressed in peripheral nociceptor neurons that initiate pain signals. One such receptor is the wasabi receptor, TRPA1, which is activated directly by a diverse panel of environmental and endogenous chemical irritants, as well as indirectly by pro-inflammatory mediators downstream of several G-protein coupled receptors, to originate painful sensations. Pain is not a single entity, but instead has many causes, types, and symptoms, including mechanical pain, temperature hypersensitivity, and pain from chronic inflammation or some chemotherapeutic agents. Despite these diversities, TRPA1 plays a central role in each etiology to generate pain signals. Indeed, two identified gain-of-function human TRPA1 natural variants, characterized by pronounced pain phenotypes in patients, provide strong evidence for the critical role of TRPA1 in human pain physiology. Accordingly, TRPA1 has been deemed a bona fide therapeutic target for analgesic and anti-inflammatory agent development, however, all known TRPA1 antagonists have failed in clinical trials due to poor pharmacokinetics. Despite its importance to human pain, little is known about how TRPA1 is regulated. A detailed understanding of the diverse mechanisms by which TRPA1 activity is modulated could uncover novel avenues for therapeutic agent development that may fare better in clinical trials.

Plasma membrane localization of TRPA1 is increased after direct channel activation or indirect activation by pro-inflammatory mediators, which could contribute to neuronal sensitization and, if unchecked, to the development of chronic pain. Nonetheless, little is known about how TRPA1 trafficking is regulated. Here, we propose to use BioID2-fused TRPA1 constructs in combination with proteomics analysis to identify interacting and proximity protein partners that govern proper channel trafficking and functionality, as our preliminary data suggests these processes are decoupled in TRPA1. In our first aim, we intend to identify protein partners necessary to mediate TRPA1 plasma membrane trafficking by using a nonfunctional TRPA1 natural variant when it is expressed alone or co-expressed with wild type protein. We recently discovered this nonfunctional natural variant exhibits aberrant trafficking alone and confers gain-of-function by assembling with wild type TRPA1 subunits to form hyperactive channels that are functional at the plasma membrane, though their trafficking route is unclear. In our second aim, we want to identify interacting and proximity protein partners necessary to confer channel functionality using wild type, functional yet N-linked glycosylation incompetent, or nonfunctional yet properly trafficking TRPA1 variants. The machinery involved in both of these processes are predicted to interact with the channel on its cytoplasmic face, which could uncover novel druggable sites that are distinct from the transmembrane regions targeted by current TRPA1 antagonists.

Phospholipase Cgamma1 in the Nucleus Accumbens Reduces Heroin-seeking: Effects on Synaptic Phospholipids and the Synaptoproteome

Ethan M. Anderson, Department of Neuroscience, Medical University of South Carolina
Chronic drug use leads to long-lasting increases in drug-seeking behavior, however, the causal molecular and cellular mechanisms responsible are not fully understood. One group of molecules that are dynamically and dramatically altered by opiate exposure and withdrawal are phospholipids of membrane bilayers including phosphatidylinositol (PI) and phosphatidylserine (PS). However, though phospholipid changes have been examined in whole striatal synaptosomes following chronic morphine exposure and withdrawal, specific changes in the nucleus accumbens (NAc) synaptosomes following heroin self-administration behavior have never been examined. One protein that regulates both phospholipids in neurons and drug-seeking behavior in the NAc is phospholipase Cgamma1 (PLCg1). I recently showed that PLCg1 overexpression in the NAc greatly reduces the motivation to take cocaine and I have unpublished preliminary data showing that endogenous NAc PLCg1 limits heroin-seeking relapse-like behaviors. However, the mechanism of how endogenous PLCg1 exerts these strong effects on drug-seeking behavior is still unknown. What is known is that PLCg1 normally cleaves phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) into inositol triphosphate (IP3) and diacylglycerol (DAG), allowing them to act as second messengers in cells. This suggests that heroin-induced changes in PLCg1 could have its effects through regulating phospholipids like PI(4,5)P2. Specifically, I hypothesize that heroin self-administration normally increases PI(4,5)P2 signaling in synapses to promote drug-seeking behavior; however endogenous PLCg1 activity reduces PI(4,5)P2 levels and therefore limits drug-seeking behavior. I also hypothesize that these effects are coupled to changes in the synaptic proteome that lead to reduced drug-seeking behavior since both PI(4,5)P2 and PLCg1 interact with many actin-binding and synaptic proteins. In this pilot grant, I propose to examine these changes using a 2x2 design in both PLCg1 knockdown and control NAc neurons from both saline and heroin self-administering rats. In Aim 1, I will determine the phosphoinositide signaling changes in NAc synaptosomes during heroin-taking, heroin withdrawal, and heroin-seeking timepoints. In Aim 2, I will determine the proteomic changes in NAc synaptosomes during heroin-seeking using the Yale/NIDA Neuroproteomics Center. These two aims will be completed using a within-subject analysis in order to correlate the phospholipid and proteomic changes in heroin-induced synapses in order to better understand the changes responsible for long-lasting heroin-seeking behaviors and the mechanism of PLCg1’s reduction of relapse-like effects.

Placenta and Neurodevelopmental Effects of in utero Cannabis Exposure

Anissa Bara, Department of Neuroscience, Icahn School of Medicine at Mount Sinai
Sociopolitical shifts in the U.S. and around the world have dramatically led to the decriminalization, medicalization, and legalization of cannabis use over the past years. The reduced risk perception of cannabis in society along with the growing wide exposure to potent cannabis (with high levels of Δ9-tetrahydrocanabinol;(THC) has contributed to it being commonly used by vulnerable groups relevant to sensitive windows of brain development such as pregnant women. Cannabinoids such as THC readily cross the placental barrier with the potential to impact the fetus. Our research group and others has accumulated evidence demonstrating that prenatal THC exposure does have long-term effects on behaviors—relevant to reward, motivation, negative affect — and molecular disturbances linked to synaptic plasticity with profound epigenetic dysregulation, especially in mesocorticolimbic brain areas. Despite the growing number of pregnant women who use cannabis and knowledge we have accrued about the long-term consequences of developmental THC exposure, there remains a dearth of information regarding biological systems downstream of such early cannabinoid perturbation that could contribute to the protracted effects especially relevant to the human condition. That question is of particular interest since it aligns with the tenet that most psychiatric disorders have a developmental genesis. Our research will fill this significant gap of knowledge by investigating an organ that is often forgotten, but which plays a crucial role during development — the placenta. We will study a unique resource of human specimens (placental and fetal brain; focus on the nucleus accumbens) to interrogate the proteome in relation to in utero cannabis exposure and subsequent offspring behavior as well as proteomic study of our translational rat model in which placenta, brain and behavior will be investigated. This study will identify early indices of psychiatric vulnerability and their developmental trajectory into adulthood.

Stress-Induced Sex- and Cell Type-Specific Proteomic Changes in the Striatum: Impact on Substance Abuse

Cheng Jiang, Department of Psychiatry, Yale University
Substance use disorder (SUD) has been identified as a major national health emergency with a devastating impact on our society and public health. Substances of abuse, such as psychostimulants and opiates, exert their reinforcing effects by inducing neural alterations in the striatum and many of these changes occur in two main subtypes of medium spiny neurons (MSNs) which are differentiated by enrichment of dopamine receptor 1 (D1) or dopamine receptor 2 (D2) and by their respective projection pathways (striatonigral D1 direct pathway and striatopallidal D2 indirect pathway). Stress is a major risk factor for the development of SUD and addiction relapse, and significantly contributes to the sex differences in vulnerability to addiction. Stress-induced addiction is thought to be mediated by dysregulated synaptic plasticity in the striatum, resulting in altered synaptic signaling in a cell type-specific manner. Therefore, understanding how stress influences the proteome of the striatum in a sex- and cell type-specific manner is a crucial first step in identifying the protein networks that underlie synaptic alterations that may precipitate addictive behaviors, and could lead to the development of novel and effective treatment options for SUD.

With the state-of-the-art technologies and expertise provided by the Yale/NIDA Neuroproteomics Center, we propose to optimize a laser capture microdissection (LCM)-based proteomics workflow to identify sex- and cell type-specific proteomic changes in D1 and D2 neurons in the striatum of mice subjected to subchronic variable stress (SCVS). In Aim 1, we will refine a method using LCM to isolate D1 and D2 neurons from the striatum of Drd1-EGFP and Drd2-EGFP mice for proteomic analysis. In Aim 2, we will identify proteomic changes in the whole striatum tissue and in striatal D1 and D2 neurons following SCVS in both female and male mice. The proposed study will allow us to optimize a LCM-based proteomics workflow and generate original data characterizing the sex differences in stress-induced cell type-specific proteomic changes in the striatum. The novel protein targets identified in our discovery-based study may also encourage future hypothesis-driven investigations into sex- and cell type-specific signaling pathways that may precipitate increased vulnerability to substance abuse.

Proteomic Characterization of κOR and Cdk5 Signaling Pathway in Rodent Nucleus Accumbens: Implications in Cocaine Addiction

Pradeep Kurup, Department of Surgery, University of Alabama at Birmingham
Addiction is a complex chronic relapsing brain disorder. Addictive drugs such as cocaine target ventral striatal circuitry to induce maladaptive changes in the intracellular signal transduction networks that are dictated by protein translational modifications by kinases and phosphatases. These neuroadaptive alterations eventually contribute to synaptic and behavioral deficits associated with drug addiction. Identifying the protein targets involved in this process will improve our current knowledge of addiction as well as impact our future therapeutic interventions. Kappa opioid receptors (κORs) are G-protein coupled receptors (GPCRs), activated by the endogenous ligand dynorphin (DYN), which elicits anti-reward or aversive-like behaviors in animals and humans. The chronic or repeated intake of cocaine progressively increases DYN levels and activates κORs as a homeostatic mechanism. κOR signaling gradually induces adaptive changes in the meso-corticolimbic circuitry, thereby reducing basal dopaminergic and glutamatergic drive on medium spiny neurons (MSNs) of the nucleus accumbens (NAc) and triggering negative emotional states during drug abstinence. These maladaptive changes caused by increased DYN/κOR signaling are one of the major contributing factors for compulsive drug craving, withdrawal symptoms, and propensity to relapse. The fundamental understanding of κOR biology is essential to unravel the critical mechanisms involved in addiction.Cdk5 is a neuronal kinase activated by its cofactor p35, which plays a prominent role in reward learning by phosphorylating several synaptic substrates. Aberrant activation of Cdk5/p35 is involved in cocaine addiction through its opposing effects on dopamine mediated PKA signaling in the NAc. In studying this phenomenon, we discovered a novel crosstalk between κOR and Cdk5 signaling in the mechanisms of cocaine addiction. Our finding shows that either chronic cocaine exposure or acute κOR agonist U50488 treatment increase the steady-state levels of p35 to cause post-synaptic activation of Cdk5 in the NAc, which is associated with decreased phosphorylation of PKA substrates. Based on these findings, we hypothesize that cocaine addiction is sustained through elevated κOR-Cdk5 signaling in the NAc. In support of this hypothesis our preliminary findings show that systemic administration of Cdk5 inhibitor (25-106) is sufficient to attenuate the cocaine-induced conditional place preference (CPP) in mice, suggesting that Cdk5 inhibition may be a potential strategy to reduce cocaine-mediated addictive behaviors. To broaden our search for identifying candidate protein targets we propose to screen the NAc proteome after chronic cocaine or acute κOR agonist (U50488) administration and compare the effects along with the 25-106 treatment. We will use affinity-based phosphoprotein substrate enrichment strategies, label-free quantitative proteomics coupled with LC/MS-MS to identify the global changes in the NAc proteome after these treatments in mice. These studies will generate novel insights on how κOR-Cdk5 signaling plays a role in cocaine addiction and will identify potential targets for further characterization.

Cell-Type-Specific Proteomic Profiling of Synaptosomes During Early and Extended Withdrawal from Self-Administered Cocaine

Yun Young Yim, Department of Neuroscience, Icahn School of Medicine at Mount Sinai
Addiction is a complex disorder that is exceptionally difficult to treat due to the high propensity for relapse even after a prolonged period of abstinence. The persistence of addiction is thought to be mediated by stable drug-induced changes in the physiology of reward-processing regions of the brain. Altered signaling within the nucleus accumbens (NAc) in particular appears to play a critical role in promoting drug-seeking and relapse. Determining these changes may reveal more effective targets to treat drug addiction and relapse. However, understanding the molecular details underlying these adaptations remains incomplete, especially in the context of cell-type and circuit specificity. Here, we propose expanding upon our previous work with the Yale/NIDA Neuroproteomics Center to examine drug-induced long-term, cell-type-specific synaptic changes within the NAc. Using wildtype, Drd1 x MT-MG, and Drd2 x MT-MG mice, we propose to profile cell-type-specific proteome changes induced by cocaine self-administration at two key time points: early (24 hours) and extended (30 days) withdrawal. After 10 days of intravenous cocaine or saline self-administration followed by 24 hours or 30 days of forced abstinence and cue-induced reinstatement test, mice will be euthanized, and the NAc will be extracted. Then, NAc synaptosomes will be purified, isolated by fluorescence-activated cell sorting (FACS), and analyzed by liquid chromatography tandem (LC-MS/MS) mass spectrometry followed by label-free quantification (LFQ) in collaboration with the Yale/NIDA Neuroproteomics Center. We anticipate characterizing alternations of synaptic proteins in a cell-type-specific manner — never before achieved — and expect to make important advances in our understanding of the molecular basis of cocaine addiction and relapse.

Capturing Altered Brain Proteomic Signaling

in Young Onset Cannabis Use Disorder in the Periphery Leveraging Label Free Proteomic Analysis of Neuron Derived Circulating Exosomes

Suhas Ganesh, Department of Psychiatry, Yale University

Background: The prevalence of cannabis use is highest amongst adolescents and young adults, who are also more vulnerable to the development of Cannabis Use Disorder (CUD) and its neuropsychiatric and cognitive consequences. This is supported by preclinical data demonstrating neuronal/synaptic pathology and immunomodulatory changes with repeated cannabinoid exposure especially in adolescents. The underlying neuropathology in humans, however, remains unclear, in part due to difficulty in obtaining brain tissues from patients with CUD. Exosomes are vesicles, that are secreted from various cell types in the brain including neurons, and which can be isolated from blood. Importantly, exosomes cross the blood brain barrier with their contents intact, making them particularly attractive vehicles for potential signatures of neuropathology in the periphery. Proteomic analyses of exosome protein cargo may help identify patterns of altered protein signaling in the brain associated with recurrent cannabis exposure. Advances in Mass Spectrometry (MS) approaches present an unprecedented opportunity for profiling proteomes in body-fluids or within organelles of interest (e.g. exosomes). MS approaches have been used to evaluate the exosomal proteome of disease states but this promising approach has not been examined in CUD. We propose to examine the proteomic changes related to recurrent cannabis exposure in plasma neuron derived exosomes (NDE) using a label-free proteomics approach to identify broad proteomic signatures of altered signaling in CUD.

Specific Aim 1: To collect plasma samples from 20 adolescents and young adults with CUD and those without any exposure to cannabis (n = 10, 5 females in each group), from the ongoing studies. Extraction of NDE from the total pool of plasma exosomes (TPE) will be done under an MTA with NeuroDex (https://www.neurodex.co/) a company with proprietary technology, ExoSORT for capturing NDE.

Specific Aim 2: To examine the broad differential proteomic profile of TPE and NDE in CUD compared to matched HCs in a label-free, discovery proteomics approach using MS. To examine the enrichment of the differentially expressed proteins to cellular components, biological processes, and molecular functions with in silico bioinformatics approaches with a focus on brain specific proteins. To specifically examine the enrichment of proteins of relevance to synaptic function using SynGo database. To validate selected proteins with western blot.

Exploratory Aim: To examine the impact of acute exposure to cannabinoids (delta-9-tetrahydrocannabinol and cannabidiol) on plasma composition and size distributions of TPEs and NDEs.

Future directions: The data collected in this pilot project will be used to support larger grant applications to NIH for examining proteomic signatures in CUD. The results of this analysis will provide valuable preliminary confirmation regarding the feasibility of deriving CNS relevant signals from the periphery in developmental neuropsychiatric disorders. This would also allow us to explore the biophysical properties of TPE and NDE in further detail in future studies. Replication of the results of this study in larger studies will have numerous translational implications for research and clinical care of persons with substance use disorders.

Investigation of the Adolescent Gut Microbiome as a Unique Driver of Protein Expression in Medial Prefrontal Cortex After Opioid Exposure

Rebecca S. Hofford, Department of Psychiatry, Icahn School of Medicine at Mount Sinai
Adolescence is a time in life when drug use is frequently initiated and use earlier in life is a predicter of problematic use during adulthood. For this reason, adolescence represents a sensitive period for the development of substance use disorder. Adolescents are undergoing many physiological changes at this time, including marked changes in the medial prefrontal cortex (mPFC), a brain region responsible for modulating motivational drive. However, changes in the brain are co-occurring with changes throughout the body. A system that is undergoing marked change during this time that is of particular interest to our group is the gut microbiome, the population of resident bacteria of the intestines that have been shown to exert effects on brain and behavior. The adolescent gut microbiome is also in flux and shifts in the predominant species of bacteria during this time have been linked to systemic inflammation, anxiety-like behavior, and stress responses. There is a growing appreciation for peripheral factors in psychiatric disturbance and our lab has shown that the gut microbiome might contribute to substance use disorder. Our preliminary results demonstrate that adolescent, but not adult, mice with their microbiome depleted with antibiotics demonstrate decreased morphine conditioned place preference after short-term gut microbiome knockdown, suggesting that adolescents are more sensitive to disruption of the gut microbiome. While there are multiple potential mechanisms of gut-brain communication, one of the best studied routes is via the production of metabolites that can cross the blood-brain barrier and affect neuronal and glial function. A number of these microbiome-derived metabolites, such as the short chain fatty acids, are known to have the ability to affect histone post-translational modifications and chromatin conformation. Given the potential of microbially-derived metabolites to alter gene expression via these epigenetic mechanisms, we performed transcriptomic profiling of adolescent and adult mPFC after manipulation of the gut microbiome. Microbiome-depleted adolescent mice treated with morphine showed massive changes in gene expression with more than a ten-fold increase in the number of regulated genes compared to morphine-treated controls with an intact microbiome. Additionally, the adolescent mice also had three-fold the amount of differentially regulated genes compared to microbiome depleted adults given morphine – suggesting that the effects of the microbiome on gene transcription are particularly potent in adolescence. However, at this point it is not known if these effects are purely transcriptional in nature or if there is a similarly large effect on protein levels. Understanding the answer to this question will be critical in dissecting out the nature of how these shifts in the microbiome alter behavioral response in translationally relevant models of substance use disorder. Here I will utilize discovery proteomics analysis in combination with drug self-administration paradigms to better understand changes in global protein expression and to clarify the underlying mechanisms of these important new findings.

Proteomics of THC-mediated changes in Extracellular Vesicle Signaling

Valeria Lallai, Neurobiology and Behavior, University of California Irvine
This project seeks to define the actions of THC on extracellular vesicle (EV) signaling in the brain. EVs have been emerging as important mediators of cell-to-cell communication throughout the brain and body. All cell types examined thus far (e.g., glia, neurons, epithelial cells, immune cells, adipose) have been found to release EVs. Under the supervision of my laboratory’s PI, Dr. Christie Fowler, we have discovered that nicotine induces the release of extracellular vesicles from cellular subpopulations in the brain. As such, we are now seeking to determine the EV-specific proteomic changes that occur following acute and/or chronic THC exposure. Our preliminary in vitro data has shown that THC administration increases cell activation and CB1 receptor mRNA expression in the choroid plexus. Given that the choroid plexus directly releases EVs into the cerebrospinal fluid (CSF), these findings support a potential interaction between cannabinoids and EV signaling. We have also shown that THC increases the expression of the EV-specific markers NCAM1 and CD63 in the CSF of adolescent male, but not female, rats. Interestingly, the NCAM1 gene encodes for neural cell adhesion molecule 1 (aka, CD56 or NCAM1), a surface marker on EVs. Of note, recent human GWAS studies have implicated allelic variation in the NCAM1 gene with cannabis use, thereby providing potential translational relevance for these studies. While we have shown these changes during adolescence, NCAM1 expression is developmentally regulated and may not be altered in adulthood with THC exposure. Therefore, in these proposed studies, adult male and female rats will be exposed to THC vapor for a single acute exposure or 14 days of chronic exposure. CSF will be collected, and EVs will be extracted for proteomic analysis. Changes in EV protein signatures will be examined, and findings may be indicative of altered release of EVs from cellular subpopulations, altered targeting of EVs to specific recipient cells, and/or altered protein cargo within EVs to induce changes in recipient cells. This project has high relevance to the Center’s theme, “Proteomics of Altered Signaling in Addiction”, as it seeks to analyze neuronal signaling mechanisms and the adaptive changes in these processes that occur in response to THC. Moreover, since EVs remain an understudied signaling mechanism within the brain, these studies will further have the potential to elucidate a novel mechanism impacting neural function that underlies drug use and abuse.

Investigating Proteins Mediating Ubiquitin-Protein Ligase Parkin-Induced Attenuation of Methamphetamine Relapse

Anna Moszczynska, Pharmacy and Health Sciences, Wayne State University
Methamphetamine (METH) use disorder (MUD) is a world-wide health problem. In the United States, more than 700,000 people abuse METH and deaths from METH overdose are rapidly rising. Despite numerous clinical trials conducted to date, there is no FDA-approved pharmacotherapy for MUD. Relapse is the biggest challenge in MUD. Preclinical studies have identified several drugs from different classes as effective against METH relapse, including glutamatergic, dopaminergic, and anti-inflammatory drugs but they have not produced desired results in those who use METH heavily. There is a clear need for novel pharmacotherapies for MUD. Novel pharmacotherapies require novel drug targets. The protein-ubiquitin ligase parkin is an attractive novel drug target for pharmacologic intervention because it is involved in several cellular mechanisms mediating relapse. We have demonstrated that young adult Park2-/- knockout rats not only self-administer more METH but also seek METH more than wild-type rats during the drug-primed relapse. Furthermore, we have demonstrated that parkin overexpression in the nucleus accumbens decreases METH self-administration. This proposal aims to determine whether overexpression of parkin in the nucleus accumbens and dorsolateral striatum decreases METH seeking during the relapse more than parkin overexpression in the nucleus accumbens alone, and which proteins and pathways are responsible for parkin effects in the nucleus accumbens vs. dorsolateral striatum. The Specific Aim 1 will determine whether male and female rats overexpressing parkin in the nucleus accumbens and dorsolateral striatum self-administer less METH during extended-access intravenous METH self-administration and during METH-induced relapse than rats overexpressing parkin only in the nucleus accumbens. The Specific Aim 2 will assess the proteomic landscape in the nucleus accumbens and dorsolateral striatum before and after METH-induced relapse. Specifically, we will first identify proteins differentially regulated by parkin overexpression in the nucleus accumbens and dorsolateral striatum. We will subsequently identify overrepresented canonical pathways, upstream regulators, protein networks, biological processes, and functions involving these proteins using bioinformatic approaches. The results from this research have a high potential to identify new protein drug targets downstream of parkin for MUD pharmacotherapy. The project is relevant to the Center’s mission because it will elucidate proteomics of altered signaling in MUD in two brain areas involved in development of cravings and relapse, and will determine which of the METH-induced proteomic changes can be rescued by parkin overexpression. These two data sets will serve as preliminary data for R01 NIH grant application for investigating proteomics of METH-altered signaling in other brain areas involved in relapse to drug use.

Identifying the PDGFRβ Signaling Pathways That Mediate Opioid Tolerance

Stephanie Puig, Department of Psychiatry, Boston University
While opioids are the gold standard for the treatment of severe pain, with continued use, opioid safety is dramatically reduced because of CNS mediated side effects, such as dependence and addiction. As a result,
addiction and death due to opioid overdose have become a national emergency. Many aspects of this opioid crisis relate to the necessity of escalating doses as tolerance develops (gradual decrease in analgesic efficacy). Thus, novel strategies and therapeutic targets are needed to increase the safety of prolonged opioid use. An ongoing challenge with regard to opioid use is how to selectively prevent tolerance, dependence and reduce addiction liability without altering their pain-relieving effect. Recent studies have shown that activation of the mu-opioid receptor (MOR) by opioids induces phosphorylation of the platelet-derived growth factor receptor beta (PDGFRβ) in the spinal cord, which is mediated by spinal release of the platelet-derived growth factor type B (PDGF-B) ligand. Accordingly, inhibition of PDGFRβ signaling with imatinib, a PDGFRβ inhibitor, or with a selective PDGF-B ligand scavenger, prevents opioid tolerance. Notably, tolerance could develop in the absence of opioids through repeated activation of PDGFRβ by the PDGF-B ligand, thus suggesting that PDGFRβ signaling specifically mediates opioid tolerance. Although PDGFRβ inhibitors are FDA approved for treatment of malignancies and could be repurposed to treat chronic pain, they also target several other receptor tyrosine kinases (RTKs), which could lead to other highly undesirable side-effects. Therefore, it is necessary to find other targets, beyond PDGFRβ signaling, that could provide safer therapeutic treatments for tolerance. New targets for tolerance could be identified through the precise understanding of the signaling cascades downstream of PDGFRβ, and through identifying proteins that are regulated by these pathways.

In this project, we propose to use proteomics to begin to understand the signaling cascades activated downstream of PDGFRβ that mediate tolerance. PDGFRβ is an RTK that possesses multiple tyrosines (Y) that when phosphorylated recruit specific signaling pathways to mediate defined cellular functions. The PDGFRβ phosphorylated tyrosines (phospho-Ys) that are phosphorylated in the context of tolerance are unknown.
Phosphoproteomics experiments could help identify the specific PDGFRβ pY activated after opioid MOR activation and would indicate which signaling cascades are recruited by PDGFRβ to initiate tolerance. Proteomics of whole cell spinal lysates would also help identify proteins that are regulated in the context of tolerance by PDGFRβ signaling. Therefore, with this funding, we plan to: 1) use phosphoproteomics to precisely determine which PDGFRβ tyrosines are phosphorylated by chronically opioid activated MORs; and 2) use proteomics to discover proteins regulated by the tolerance inducing PDGFRβ signaling activated by chronic opioid treatments. Our laboratory will benefit from a proteomics approach, as this high throughput method will enable us to precisely identify PDGFRβ signaling cascades that mediate tolerance. These experiments will lay the foundation for future experiments and grant proposals that will focus on identifying targets that could help preserve long term opioid analgesic efficacy and prevent the occurrence of dependence and addiction.

Protein Analysis from Organelles and Cellular Compartments of Neuronal Populations

Yotam Sagi, Laboratory of Molecular and Cellular Neuroscience, Rockefeller University
Changes in protein content within cellular organelles and subcellular compartments are essential in mediating the action of drugs of abuse and their treatments. Our understanding of drug addiction is limited by the lack of methods that enable isolation of proteins from cellular compartments in neuronal sub-populations. We overcome this by utilizing mice that express tagged ribosomes in defined neuronal types. Using recently-modified protocols for fluorescent-activated sorting or synaptosomal preparations followed by immunoprecipitation, we will isolate proteins from cell nuclei and synaptic compartments of hippocampal inhibitory interneurons and excitatory granule cells. Using the cocaine conditioned place preference paradigm, our study will identify cell-type specific subcellular changes associated with the extinction process.

Profiling Microbial Metabolites To Elucidate The Relationship Between SCFAs, Intestinal Permeability, and Alcohol

Summer L. Thompson, Department of Psychiatry, Yale University
The community of bacteria populating the gut, the gut microbiota, recently emerged as a major modulator of neuropsychiatric phenotypes. The gut microbiota are implicated in several substance use disorders including alcohol use disorder. Alcohol consumption alters the gut microbiota, the population of bacteria that colonize the gastrointestinal tract. One study found that altered gut microbiota in abstinent individuals with alcohol use disorder predicted worse withdrawal symptoms and likelihood of relapse. The same individuals showed greater intestinal permeability, a phenotype that is also modulated by the gut microbiota and can lead to systemic inflammation. This finding suggests that intestinal permeability may also play a role in behavioral aspects of alcohol consumption, consistent with the neuroinflammatory effects of alcohol. Evidence in rodents suggests that differences in the types of bacteria populating the gut can predict alcohol consumption and related gene expression in the brain. Dietary changes, fecal microbial transplantation, or antibiotic administration have been reported to modulate alcohol consumption or withdrawal symptoms in rodents. Prebiotics are a type of fiber that “feeds” beneficial bacteria in the gastrointestinal tract, promoting healthy microbiota. Incorporation of prebiotics into the diet protects against alcohol-induced changes in the microbiota and leakage of bacteria from the gastrointestinal tract that leads to systemic inflammation. Recent work found that prebiotics reduced signs of withdrawal from chronic alcohol consumption in rats, suggesting that prebiotics could influence behavioral consequences of chronic alcohol consumption. Indeed, prebiotics ameliorate stress-induced anxiety- and depression-like behavior and gene expression in the brain of rodents. However, there are multiple candidate mechanisms for a role of prebiotics in alcohol use behaviors. One possibility is that prebiotic-induced fortification of the intestinal lining reduces downstream neuroimmune effects, which are heavily implicated in alcohol use behaviors. Another possibility is that prebiotics facilitate production of short-chain fatty acids (SCFAs) that enter circulation and affect central signaling directly. SCFAs are metabolites produced by the beneficial bacteria that increase in growth by fermenting prebiotics. SCFAs play a causal role in maintaining the lining of the gastrointestinal tract and protecting it from alcohol-induced injury. Furthermore, SCFA levels in the gut are reduced in chronic alcohol users and are restored by fecal transplant associated with decreased alcohol craving, but SCFA levels in the brain following alcohol intake have not been documented. Numerous studies have documented behavioral effects of SCFAs, which are known to cross the blood-brain barrier and be psychoactive, but the exact mechanisms of these effects remain incompletely understood. In addition, measuring SCFA levels can be challenging; SCFA levels are sparse in the brain, whereas fecal samples are highly complex in combination with the low molecular mass of SCFAs, together requiring innovative approaches for accurate measurement. These findings highlight the need for a better understanding of the role of SCFAs in alcohol use. Here, we seek to determine how prebiotics alter SCFA levels in the gut and brain in mice that chronically self-administered alcohol.