Abstract Objective Fibroblast growth factor 21 (FGF21) analogs have been tested as potential therapeutics for substance use disorders. Prior research suggests that FGF21 administration might affect alcohol consumption and reward behaviors. Our recent report showed that plasma FGF21 levels were positively correlated with alcohol use in patients with alcohol use disorder (AUD). FGF21 has a short half-life (0.5–2 h) and crosses the blood–brain barrier. Therefore, we set out to identify molecular mechanisms for both the naïve form of FGF21 and a long-acting FGF21 molecule (PF-05231023) in induced pluripotent stem cell (iPSC)-derived forebrain neurons. Methods We performed RNA-seq in iPSC-derived forebrain neurons treated with naïve FGF21 or PF-05231023 at physiologically relevant concentrations. We obtained plasma levels of FGF21 and GABA from our previous AUD clinical trial (n = 442). We performed ELISA for FGF21 in both iPSC-derived forebrain neurons and forebrain organoids. We determined protein interactions using co-immunoprecipitation. Finally, we applied ChIP assays to confirm the occupancy of REST, EZH2 and H3K27me3 by FGF21 using iPSC-derived forebrain neurons with and without drug exposure. Results We identified 4701 and 1956 differentially expressed genes in response to naïve FGF21 or PF-05231023, respectively (FDR < 0.05). Notably, 974 differentially expressed genes overlapped between treatment with naïve FGF21 and PF-05231023. REST was the most important upstream regulator of differentially expressed genes. The GABAergic synapse pathway was the most significant pathway identified using the overlapping genes. We also observed a significant positive correlation between plasma FGF21 and GABA concentrations in AUD patients. In parallel, FGF21 and PF-05231023 significantly induced GABA levels in iPSC-derived neurons. Finally, functional genomics studies showed a drug-dependent occupancy of REST, EZH2, and H3K27me3 in the promoter regions of genes involved in GABA catabolism which resulted in transcriptional repression. Conclusions Our results highlight a significant role in the epigenetic regulation of genes involved in GABA catabolism related to FGF21 action. (The ClinicalTrials.gov Identifier: [47]NCT00662571) Keywords: FGF21, GABA catabolism, Alcohol use disorder Highlights * • FGF21 analogs may have utility as therapeutics for substance use disorders. * • FGF21 was positively correlated with GABA levels in patients with AUD. * • FGF21 increased GABA levels in iPSC-derived neurons and brain organoids. * • FGF21 mediated the occupancy of REST, EZH2 and H3K27me3. * • The REST-EZH2-H3K37me3 complex mediates gene repression in a drug dependent manner. Abbreviations FGF21 Fibroblast growth factor 21 AUD Alcohol use disorder iPSC Induced pluripotent stem cell GABA γ-aminobutyric acid EB Embryoid body RIN RNA integrity numbers FDR False discovery rate GSEA Gene Set Enrichment Analysis DAVID The database for Annotation, Visualization and Integrated Discovery TF Transcription factor ELISA Enzyme-linked immunosorbent assay co-IP Co-immunoprecipitation ChIP Chromatin immunoprecipitation REST Repressor element 1 silencing transcription factor TAF2 TATA-box binding protein associated factor 2 H3K27me3 Tri-methylation of lysine 27 on histone H3 protein 1. Introduction Alcohol use disorder (AUD) is the most prevalent substance use disorder worldwide [[48]1]. To date, acamprosate, naltrexone, and disulfiram have received United States Food and Drug Administration approval for the treatment of AUD. However, growing evidence has prompted the development of fibroblast growth factor 21 (FGF21) based therapy as a potential treatment for AUD [[49][2], [50][3], [51][4]]. A major goal of AUD research is the development and optimization of effective drugs to treat AUD. Over the past three decades, clinical trials for AUD have been conducted on more than 30 different drugs [[52]5]. However, most clinical trials showed no significant impact or the effect size was small [[53]5]. This may be true, at least in part due to 1) the heterogeneity of AUD phenotypes, and 2) the critical knowledge gaps underlying the mechanisms of action of the medications used to treat AUD and the pathophysiology of AUD. FGF21 analogs have been tested in clinical trials for metabolic diseases, i.e. type 2 diabetes and non-alcoholic fatty liver disease, and they are generally well-tolerated [[54]6]. FGF21 is a peptide hormone that is primarily secreted by the liver, but it also crosses the blood–brain barrier [[55]7]. Prior literature suggests that FGF21 administration could affect alcohol use and reward behaviors [[56]3]. Our recent study showed that FGF21 levels were associated with recent alcohol consumption in patients with AUD [[57]8]. Specifically, we observed a positive correlation between plasma FGF21concentrations and recent alcohol consumption, ie the number of drinks per day in the past 30 days and 90 days [[58]8]. Furthermore, we demonstrated that the effects of FGF21 on induced pluripotent stem cell (iPSC)-derived 3D-brain organoids were associated, at least in part, with the regulation of catecholamines—neurotransmitters that are modulated by γ-aminobutyric acid (GABA) neurotransmission and which play a crucial role in reward pathways [[59]2,[60]8]. These observations potentially open the way to pharmacologic manipulation of FGF21, which in turn could have significant implications for the regulation of neurotransmitters and alcohol use. GABA is the primary inhibitory neurotransmitter in the brain, and it has been implicated in substance use disorders [[61]9]. Research has shown that changes in GABA concentrations in the prefrontal cortex can lead to deficits in decision-making and impulse control, which may contribute to the development of addiction [[62]10]. However, GABA may have a complex role in these pathways that is not fully understood at present [[63]9]. Current in vitro models for studying drug action in the human brain are limited. Therefore, we used iPSC-derived neurons as a novel “cell-line based” approach to advance our understanding of drug action in the brain. Since the half-life of the naïve form of FGF21 is about 0.5–2 h [[64]11], it may not be suitable for clinical use due to poor pharmacokinetic and biophysical properties [[65]12]. Therefore, we designed the present study to investigate both the naïve form of FGF21 and a long-acting FGF21 molecule (PF-05231023), which was created by conjugating two molecules of modified FGF21 (dHis/Ala129Cys) to an antibody scaffold [[66]13]. We set out to identify molecular mechanisms for the naïve form of FGF21 and a long-acting FGF21 molecule (PF-05231023) in iPSC-derived neurons. We began by determining the gene expression profiles of these two compounds in iPSC-derived forebrain neurons. We found remarkable overlap in both up-regulated and down-regulated genes between the naïve form of FGF21 and PF-05231023. We then further investigated these overlapping genes and identified pathways enriched for genes involved in GABA catabolism. Finally, we identified a possible epigenetic mechanism related to the regulation of GABA catabolism by FGF21. These studies have not only extended our original findings with regard to FGF21 and catecholamine metabolism [[67]8], but they have also revealed a novel mechanism related to a pharmacologic action of FGF21 in GABA catabolism. 2. Methods 2.1. Study subjects and ethics statements This study was conducted under protocols (20-00372 and 18-006428) reviewed and approved by the Mayo Clinic Institutional Review Board. We maintained confidentiality for all study participants. All participants whose samples were used in this study gave their consent for participation in the study and for publication of the study results in a peer-reviewed scientific journal. 2.2. Generation of iPSCs, iPSC-derived forebrain neurons, and 3-D brain organoids Induced pluripotent stem cells (iPSCs) were generated from four subjects with alcohol use disorder (two males and two females), see [68]Supplementary Table 1. Specifically, we performed iPSC reprogramming using peripheral blood mononuclear cells (PBMCs) and the CytoTune™-iPS 2.0 Sendai Reprogramming Kit (A16517, Thermo Fisher, USA) as described previously [[69]14,[70]15]. All iPS cell lines showed normal karyotypes, and they all expressed pluripotency markers. Cell lines were regularly characterized and verified to be free from mycoplasma. We then generated iPSC-derived forebrain neurons as reported previously [[71]14,[72]16]. Briefly, iPS cells were cultured on Matrigel with mTeSR1 Plus media (STEMCELL technology, MA, USA). We suspended 3-D iPSC aggregates in embryoid body (EB) medium, consisting of FGF2-free iPS cell medium supplemented with 2 μM dorsomorphin (STEMCELL technology, MA, USA) and 2 μM A-83 (STEMCELL technology, MA, USA), in non-treated polystyrene plates for 6 days. EB medium was renewed every day. After 6 days, we replaced EB medium by neural induction medium (NPC medium) consisting of DMEM/F12, 1× N2 supplement, 1× NEAA, 2 μg ml^−1 heparin (Tocris Bioscience) and 2 μM cyclopamine (Tocris Bioscience). The floating EBs were then transferred to Matrigel-coated plates at day 7 to form neural tube-like rosettes. The attached rosettes were kept for 15 days with NPC medium change every other day. On day 22, we transferred the rosettes to low attachment plates in NPC medium containing 1× B27 (Thermo Fisher). On day 24, neural progenitor spheres were dissociated and placed onto Matrigel-coated plates in neuron culture medium, consisting of Neurobasal medium supplemented with 1× glutamax, 1× B27, 0.2 mM l-Ascorbic Acid, and 0.5 mM cAMP (Sigma-Aldrich, St Louis, MO), 10 ng/ml BDNF and 10 ng/ml GDNF. Medium was replaced every three days during continuous culture [[73]17]. The inhibitory/excitatory subtype classification was characterized by immunocytochemistry ([74]Supplementary Figure 1). Specifically, four weeks after differentiation from NPC, most neurons expressed a glutamatergic marker (VGLUT1). Approximately 5–10% of neurons are GABAergic neurons that express glutamate decarboxylase 1 (GAD1), and less than 1% of the neurons are dopaminergic neurons that express tyrosine hydroxylase (TH). The iPSC-derived neurons functionally mature four weeks after differentiation from NPCs. For example, they form synapses, fire action potentials, and have spontaneous synaptic activity [[75]17]. Neurotransmitters including GABA were also detected in the iPSC-derived neurons [[76]18] ([77]Supplementary Figure 2A). Drug treatment for iPSC-derived forebrain neurons was initiated four weeks after differentiation from NPC. We also generated 3-D iPSC-derived forebrain organoids. Briefly, EBs were embedded in matrigel and cultured with 1× N2, 1× NEAA and 1× Glutamax (Invitrogen, Grand Island, NY), 1 μM SB431542, and 1 μM CHIR99021 (Selleckchem, Carlsbad, CA) for seven days. On day 14, organoids were mechanically dissociated from the matrigel and cultured in a bioreactor [[78]19]. Culture medium from days 14–70 consisted of DMEM/F12 medium supplemented with 1× N2, 1× B27, 1× NEAA and 1× Glutamax, 1× 2-metabptoethanol, 100× penicillin–streptomycin solution, and 2.5 μg/ml insulin (Sigma-Aldrich, St Louis, MO). The medium was changed every other day. From day 70 onward, supplementing media with 20 ng/ml BDNF, 20 ng/ml GDNF (Peprotech, Rocky Hill, NJ), 0.2 mM l-Ascorbic Acid, and 0.5 mM cAMP (Sigma-Aldrich, St Louis, MO) was used [[79]20]. Neurotransmitters i.e. GABA, serotonin, and glutamate, were also detected in the iPSC-derived forebrain organoids [[80]18] ([81]Supplementary Figure 2B). Our recent single cell RNA-seq in iPSC-derived forebrain organoids suggested that the number of GABAergic cells is ∼28% of the total neuronal population at day 90 [[82]16]. Drug treatment for iPSC-derived forebrain organoids was initiated from day 83 to day 90 [[83]16]. 2.3. Drug treatment The naïve form of FGF21 (100 ng/ml, PeproTech, cat: 100-42) at a physiologically relevant concentration was used to perform the proposed experiments [[84]21]. Concentrations of the naïve form of FGF21 (Peprotech, cat#100-42) [[85]22,[86]23] and PF-05231023 (MedChemExpress, cat: HY-113697) [[87]24,[88]25] were selected to fall within the range of blood concentrations for these drugs observed during preclinical and clinical studies. We treated cells with either the naïve form of FGF21 or PF-05231023 for 7 days, with a daily medium change. 2.4. Immunostaining and confocal microscopy Cells were fixed in paraformaldehyde (4%) at room temperature for 15 min. Cells were permeabilized with Triton X-100 (0.2%) in PBS. After blocking, cells were incubated with the primary antibody (see [89]Supplementary Table 1) in 5% BSA overnight. After a washing step, they were then incubated for 1 h at room temperature with secondary antibodies. Antifade mounting media with dapi (VECTOR laboratory, Burlingame, CA, USA) was used to stain the nuclei. Fluorescence microscopy (Olympus, FV1200) was used to visualize slides. 2.5. RNA sequencing and data analysis Total RNA was extracted using Trizol and the RNeasy mini kit (Qiagen, Valencia, CA, USA). The RNA integrity numbers (RIN) were above 9.5 for RNA samples. We performed RNA-seq with four AUD patients and each subject included three treatment conditions (vehicle, naïve FGF21 and PF-05231023) and two technical replicates. RNA-seq experiments were conducted by GENEWIZ using an Illumina HiSeq 6000 with eight samples in each lane using 100 bp paired end index reads ([90]Supplementary Table 2). Fastq files containing paired RNA-Seq reads were aligned with STAR [[91]26] against the UCSC human reference genome (hg19). RNA-seq differential expression analysis was performed using the DESeq2 package with default parameters [[92]27]. The Wald test was used to compare the beta estimate divided by its estimated standard error to a standard normal distribution to derive p-values. P values were then adjusted for multiple testing using the Benjamini and Hochberg method to produce false discovery rate (FDR) values. A false discovery rate less than 0.05 was considered statistically significant. Gene Set Enrichment Analysis (GSEA) software [[93]28,[94]29], and the Database for Annotation, Visualization and Integrated Discovery (DAVID) [[95]30,[96]31] were used to perform pathway enrichment analysis. We then performed over-representation analysis [[97]32] for transcription regulatory targets of individual transcription factor (TF) using the Fisher's exact test method for selected gene lists i.e. the top 500 or 200 genes against the transcription factor target gene sets. Specifically, target genes of transcription factors determined from transcription factor binding site profiles were downloaded from ENCODE [[98]33,[99]34], and MSigDB [[100]35]. The R function fisher.test with parameter alternative = “greater” was used to determine whether selected gene sets were significantly over-represented in the gene lists. 2.6. Enzyme-linked immunosorbent assay (ELISA) for GABA We measured GABA levels in the culture medium of iPSC-derived neurons and iPSC-derived brain organoids with the human GABA ELISA kit (Abcam: [101]AB287792) in accordance with the manufacturer's instructions. Specifically, we used 100 μL of culture medium for ELISA. We made an 8-point standard concentration curve using the calibration standard solution to quantify GABA concentrations. ELISA was performed in duplicate and absorbances were read on a microplate reader at 450 nm. 2.7. Co-immunoprecipitation (co-IP) and Western blot analysis We used iPSC-derived neurons for co-IP. Cells were resuspended in 500 μL IP lysis buffer containing 5 μL protease inhibitor cocktail and were incubated on ice for 30 min. Anti-REST (Proteintech, Rosemount, IL, USA) and anti-EZH2 antibodies and control IgG (Cell Signaling Technology, Danvers, MA, USA) were used to perform IP. Specifically, IP Samples containing protein A/G agarose beads (ThermoFisher, Pierce™ Protein A agarose Beads, cat#:20333, USA) were rotated at 4 °C overnight. Immunoprecipitates were washed three times with ice cold lysis buffer. Protein samples were eluted with 100 μL 2× Laemmli loading buffer and boiled at 95 °C for 10 min. Proteins were separated on 4–12% SDS-PAGE gels and transferred onto PDVF membranes. After blocking, membranes were incubated with primary antibodies against REST or EZH2 at 4 °C overnight. The washed membrane was then incubated with secondary antibody for an hour at room temperature. The membranes were then incubated with super signal ECL substrate (Thermo Scientific, Madison, WI, USA) and were visualized with the Geldoc Go Gel imaging system (Bio-rad, USA). Antibodies for co-IP and Western blot analysis are listed in [102]Supplementary Table 1. 2.8. Chromatin immunoprecipitation (ChIP) assay ChIP assays were performed using iPSC-derived neurons and the MAGnify™ Chromatin Immunoprecipitation System (ThermoFisher, cat#492024, USA). REST, EZH2, and H3K27me3 complexes were immunoprecipitated using antibodies against REST, EZH2, and H3K27me3, respectively. Control IgG was used (Cell Signaling Technology, Danvers, MA, USA) as a negative control. Real time PCR was used to quantify DNA-protein binding. Antibodies and primers for ChIP assays are listed in [103]Supplementary Table 1. 2.9. Statistical analysis Statistical analyses were performed using R Statistical Software (version 4.2.2; R Foundation for Statistical Computing, Vienna, Austria). The correlation between plasma FGF21 levels and plasma GABA levels was tested using the Spearman rho correlation coefficient. ELISA and ChIP assays were analyzed using ANOVA, followed by Tukey's multiple comparison tests for individual comparisons when significant effects were detected. P < 0.05 was considered statistically significant. 2.10. Data availability All data supporting our findings can be found in the main paper or in supplementary files. Sequencing data are available via the GEO accession number: [104]GSE223173. 3. Results 3.1. Gene expression profiles in iPSC-derived neurons treated with FGF21 or a long-acting FGF21 molecule (PF-05231023) The present study investigated the molecular mechanisms of the naïve form of FGF21 and PF-05231023 using iPSC-derived forebrain neurons. Previous preclinical and clinical studies reported vital involvement of the prefrontal cortex in substance use disorders [[105]36,[106]37]. As a result, we generated iPSCs using peripheral blood mononuclear cells from four unrelated individuals (2 men and 2 women) and differentiated them into forebrain neurons ([107]Figure 1). We treated these iPSC-derived forebrain neurons with the naïve form of FGF21 (100 ng/ml) [[108]22,[109]23] and PF-05231023 (20 μM) [[110]24,[111]25] with concentrations falling within the range of blood concentrations for these drugs observed during preclinical and clinical studies. We identified 4701 and 1956 genes that displayed significant changes in expression after the cells were treated with either naïve FGF21 or PF-05231023, respectively (FDR < 0.05), as determined by RNA-seq ([112]Figure 2A). The effects of FGF21 on the central nervous system have been associated with the regulation of catecholamines [[113]2,[114]8]. In line with those observations, we identified a series of biological pathways i.e., neurotransmitter receptor activity and catecholamine secretion, that were the most common and most highly affected pathways in response to naïve FGF21 or PF-05231023 ([115]Supplementary Tables 3 and 4). Furthermore, we found that a large number of differentially expressed genes in response to naïve FGF21 or PF-05231023 exposure overlapped ([116]Figure 2B). Notably, those overlapping genes displayed identical gene expression patterns after drug treatment ([117]Figure 2C and [118]Supplementary Table 5). Furthermore, when we used those overlapping genes for pathway analysis, we found that the GABAergic synapse pathway was the most significant biological pathway identified ([119]Supplementary Table 6). We then performed pathway analysis for up-regulated or down-regulated genes separately ([120]Supplementary Tables 7 and 8). Notably, the GABAergic synapse pathway was the most significant pathway affected by the down-regulated genes ([121]Supplementary Table 7). These results significantly expanded our original observations with regard to the role of FGF21 in catecholamine metabolism [[122]8], and served to highlight a possible novel mechanism of FGF21 or the long-acting FGF21 molecule (PF-05231023). Therefore, the next series of studies was designed to 1) determine the association of FGF21 and GABA in the plasma samples from patients with AUD, and 2) to identify possible molecular mechanisms related to this relationship. Figure 1. [123]Figure 1 [124]Open in a new tab Generation of iPSC-derived neurons. (A) iPSC generation using peripheral blood mononuclear cells (PBMC). All iPSC cell lines displayed normal karyotypes. The panel below the schematic displays representative examples of staining for iPSC pluripotency markers: SOX2, and TRA-1-81. All iPSC lines were positive for pluripotency markers. (B) A schematic outline of procedures used during the differentiation of iPSC-derived forebrain neurons. The panel below the schematic displays representative examples of staining for TUJ1 and MAP1/2. Figure 2. [125]Figure 2 [126]Open in a new tab Bulk RNA-seq using iPSC-derived forebrain neurons treated with FGF21, PF-05231023 and vehicle. (A) Volcano plots indicate differentially expressed genes with FDR 0.05. (B) Venn diagram showing expression profiles for the 974 genes with expression that could be affected by both FGF21 and PF-05231023 as determined by RNA-seq (FDR < 0.05). The middle and bottom Venn diagrams illustrate overlapping up-regulated and down-regulated genes in response to FGF21 and the long-acting FGF21 molecule. Those results indicate that those overlapping genes displayed identical directionality. (C) Heatmap showing expression profiles for the 974 genes affected by FGF21 and PF-05231023. 3.2. FGF21 was positively correlated with GABA concentrations in patients with AUD We previously conducted a clinical trial with a total of 442 patients with AUD. FGF21 and GABA were assayed using plasma samples as described previously [[127]8,[128]20]. We collected plasma samples from AUD patients prior to acamprosate treatment. We observed a significant positive correlation of FGF21 and GABA concentrations in the plasma samples of patients with AUD (r: 0.13, p: 0.008) ([129]Figure 3A). Since FGF21 can cross the blood brain barrier [[130]7], this led us to test whether FGF21 might modulate levels of GABA in iPSC-derived brain cells. As anticipated, the concentrations of GABA in the culture medium increased significantly in response to FGF21 treatment ([131]Figure 3B). In line with this finding, when iPSC-derived forebrain neurons were exposed to PF-05231023, the concentrations of GABA in the culture medium also increased substantially ([132]Figure 3B). We next generated iPSC-derived forebrain organoids to verify those results ([133]Figure 4A). Brain organoids derived from iPSCs are three-dimensional self-assembled structures containing multiple brain cell types such as neurons, astrocytes, and microglia [[134]38,[135]39], which can mimic human brain structures and which might help to bridge the gap between preclinical and clinical trials. Consistently, the levels of GABA in the culture medium of iPSC-derived forebrain organoids increased significantly in response to either naïve FGF21 or PF-05231023 treatment ([136]Figure 4B). Our findings suggest that both naïve FGF21 and PF-05231023 might play a role in the regulation of GABA catabolism. Therefore, we designed the next series of studies to determine the molecular mechanism mediating this relationship. Figure 3. [137]Figure 3 [138]Open in a new tab Plasma FGF21 concentrations were positively correlated with plasma GABA concentrations. (A) Plasma FGF21 concentrations were associated with plasma GABA levels in patients with alcohol use disorder. (B) both FGF21, and PF-05231023 could substantially increase the level of GABA in the culture medium of iPSC-derived forebrain neurons. Figure 4. [139]Figure 4 [140]Open in a new tab FGF21 and PF-05231023 induced GABA concentrations in iPSC-derived forebrain organoids. (A) A schematic outline of procedures used during the differentiation of iPSC-derived forebrain organoids. The middle panel displays images of iPSC-derived forebrain organoids during differentiation. The bottom panel of the schematic displays representative examples of staining for neural progenitors (SOX2) and neurons (TUJ1). (B) FGF21 and PF-05231023 substantially induced the concentrations of GABA in the culture medium of iPSC-derived forebrain organoids. 3.3. REST, EZH2 and H3K27me3 co-occupied the promoter regions of genes associated with GABA catabolism We performed upstream regulator analysis using selected gene sets, i.e., the threshold of the top 500 down-regulated or up-regulated genes in the presence of naïve FGF21 or PF-05231023 ([141]Figure 2A) [[142]32]. That analysis allowed us to identify the cascade of upstream transcription regulators that might contribute to gene expression pattern changes in response to drug treatment. Repressor element 1 silencing transcription factor (REST) was predicted to be the most important upstream regulator of the genes down-regulated in response to naïve FGF21 or PF-05231023 treatment with p values of 9.14E-36 and 1.04E-47 respectively ([143]Supplementary Table 9). This trend remained when we set the threshold of the top 200 down-regulated genes ([144]Supplementary Table 9). In contrast, TATA-box binding protein associated factor 2 (TAF2) was predicted to be the most important upstream regulator of genes up-regulated in response to FGF21 or PF-05231023 treatment, with p values of 8.21E-17 and 6.06E-09, respectively ([145]Supplementary Table 9). Notably, 14 out of 15 genes that were associated with the GABAergic synapse pathway ([146]Supplementary Table 6) were down-regulated in the presence of naïve FGF21 or PF-05231023 as determined by RNA-seq ([147]Figure 5A). This observation led us to ask whether REST might play a role in transcription regulation for the 15 genes associated with GABA catabolism. We began by exploring the ENCODE database and found that REST could bind to the promoter regions of all of those genes except GABRA3 and CACNA1C. As a result, we performed ChIP assays to confirm the occupancy of REST on the promotors of those genes in iPSC-derived forebrain neurons ([148]Figure 5B). Previous studies have shown that EZH2 is a co-factor for REST [[149]40]. Of importance, EZH2 was one of the top upstream regulators of genes that were down-regulated in the presence of naïve FGF21 and PF-05231023 ([150]Supplementary Table 9). We also performed co-IP to demonstrate whether REST could physically interact with EZH2 in iPSC-derived neurons ([151]Figure 5C). EZH2 is a catalytic subunit of polycomb repressive complex 2 (PRC2) which catalyzes the tri-methylation of lysine 27 on the histone H3 protein (H3K27me3) to regulate gene expression through epigenetic machinery [[152]41]. Specifically, H3K27me3 is a transcriptionally repressive histone mark, and it can function as a silencer to repress gene expression [[153]42]. We then performed ChIP assays to show that REST, EZH2 and H3K27me3 were able to bind to the same DNA regions in iPSC-derived forebrain neurons ([154]Figure 5B). These results confirmed the co-occupancy of REST, EZH2 and H3K27me3. However, they did not explain drug-induced inhibition of gene expression when the cells were treated with FGF21 and PF-05231023 ([155]Figure 5A). Figure 5. [156]Figure 5 [157]Open in a new tab REST, EZH2 and H3K27me3 co-occupied the promoter regions of genes associated with GABA catabolism. (A) Heatmap showing expression profiles for the 15 genes involved in the GABAergic synapse pathway. (B) ChIP assays showing that REST, EZH2 and H3K27me3 co-occupied the promoter regions of the 13 selected genes as shown in [158]Figure 5A that were associated with the GABAergic synapse pathway in response to FGF21 and PF-05231023 treatment of iPSC-derived forebrain neurons (n = 4). Percentages of ChIP DNA/input were determined by qPCR. Data are represented as % input, (enrichment relative to IgG control) = % input (antibody against REST, EZH2 and H3K27me3) – % input (IgG). ChIP-qPCR was performed in triplicate. Three independent experiments were performed. This figure represents results from one of the three experiments. (C) Co-Immunoprecipitation (IP) was used to determine whether REST protein could interact with EZH2 in iPSC-derived forebrain neurons. Whole cell lysates from iPSC-derived forebrain neurons were immunoprecipitated with anti-REST antibodies or anti-IgG antibodies and protein samples were immunoblotted and probed with antibodies against REST or EZH2. 3.4. REST-EZH2-H3K37me3 complex mediated gene repression in a drug-dependent fashion It is well-documented that transcription factors (TF) bind to specific DNA binding motifs to regulate gene expression in a drug-dependent fashion [[159][43], [160][44], [161][45]]. We next performed ChIP assays using iPSC-derived forebrain neurons after exposure to naïve FGF21 or PF-05231023. When cells were treated with naïve FGF21 or PF-05231023, the vast majority of the genes tested, including ABAT, a gene encoding a key enzyme responsible for GABA catabolism, GRBBR1 (GABA B receptor 1), and SLC32A1 (a GABA transporter), displayed significantly increased REST binding ([162]Figure 6). As mentioned previously, REST is also known as neuron-restrictive silencer factor, a protein that is encoded by the REST gene and which acts as a transcriptional repressor. Thus, changes in downstream gene expression could be mediated by the binding affinity of REST [[163]46]. We had demonstrated that REST co-exists with EZH2 and H3K27me3 in the target promoters in iPSC-derived forebrain neurons ([164]Figure 5B). EZH2 is the catalytic unit of the polycomb repressive complex 2 (PRC2) with methyltransferase activity. PRC2 is essential for transcription silencing by specifically catalyzing the formation of H3K27me3, a marker of transcriptional repression [[165]42]. We next attempted to determine whether the recruitment of EZH2 and H3K27me3 could be altered in a drug-dependent manner. Those results might explain repression of the list of genes involved in GABA catabolism, as shown in [166]Figure 5A. As anticipated, for the same genes (see [167]Figure 6), the recruitment of EZH2 and H3K27me3 was significantly induced by naïve FGF21 or PF-05231023. Figure 6. [168]Figure 6 [169]Open in a new tab REST-EZH2-H3K37me3 complex-mediated gene repression in a drug dependent fashion. ChIP assays were performed in iPSC-derived forebrain neurons treated with FGF21, PF-05231023 and vehicle. We confirmed that changes in REST binding were correlated with changes in mRNA expression for all 13 genes. Specifically, in the presence of FGF21, all 13 binding sites except GNAI1 displayed significantly increased REST binding. In parallel, the recruitment of EZH2 and H3K27me3 also increased substantially, thus confirming that changes in REST-EZH2-H3K37me3 occupancy were highly correlated with transcription. Taken together, our results showed that FGF21 could increase GABA concentrations in both iPSC-derived forebrain neurons and forebrain organoids ([170]Figure 3, [171]Figure 4B). GABA is synthesized primarily from glutamate by glutamate decarboxylases, GAD1 and GAD2, in presynaptic neurons. The GABA synthesized is loaded into synaptic vesicles via a vesicular GABA transporter (VGAT also known as SLC32A1). GABA activity is terminated at the synapse by uptake into both nerve terminals and surrounding cells via plasma member GABA transporters. GABA is then catabolized by GABA transaminase (ABAT) [[172]47]. Most of the genes related to GABA catabolism can be suppressed by naïve FGF21 or PF-05231023. This might be, at least in part, because FGF21 and PF-05231023 can facilitate the recruitment of REST-EZH2-H3K37me3 to the promotors of target genes, which in turn, represses gene expression ([173]Figure 7). Figure 7. [174]Figure 7 [175]Open in a new tab Schematic diagram illustrating the effects of FGF21 on GABA catabolism. Specifically, FGF21 and PF-05231023 significantly induced GABA levels in iPSC-derived forebrain neurons. We identified a drug-dependent occupancy of REST, EZH2, H3K27me3 in the promoter regions of genes associated with GABA catabolism i.e., ABAT (a key enzyme responsible for GABA catabolism), GRBBR1 (GABA B receptor 1), and SLC32A1 (GABA transporter), which resulted in transcriptional repression. 4. Discussion Substance use disorders such as AUD are complex and multifaceted disorders. Current FDA-approved treatment options are limited and have limited efficacy [[176]48]. FGF21 has been identified as a potential therapeutic target for substance use disorders due to its ability to regulate reward pathways in the brain [177][3], [178][49]. As a result, studying FGF21's mechanism of action is critical as it potentially opens a new avenue for developing more effective and safer treatments for addiction. Like most neuropsychiatric disorders, addiction is highly heterogeneous. This makes it challenging to develop models or tools that can capture the full range of individual differences in addiction. However, breakthroughs in iPSC technology offer a powerful research tool for studying neuropsychiatric disorders and can potentially revolutionize the field of neuroscience by providing new insights into disease mechanisms and by facilitating the development of new drugs. Our recent study demonstrated that plasma FGF21 levels were positively associated with recent alcohol use in patients with AUD, and that ethanol can induce the concentrations of FGF21 and catecholamines in iPSC-derived brain organoids [[179]8]. FGF21 has been suggested as a potential therapeutic target for the treatment of addictions. The role of FGF21 in metabolism has been studied extensively, however, molecular mechanism(s) underlying FGF21's mechanism of action in the brain remain unclear. As a result, the present study was designed to investigate the molecular mechanisms of naïve FGF21 and the long-acting FGF21 compound (PF-05231023) using iPSC-derived forebrain neurons, and the omics data from our previous AUD clinical study. The present study identified a large number of differentially expressed genes in response to exposure to naïve FGF21 or PF-05231023. Specifically, 974 differentially expressed genes overlapped between treatment with naïve FGF21 and with PF-05231023. REST was predicted as the most significant upstream regulator of differentially expressed genes in the presence of naïve FGF21 or PF-05231023. The GABAergic synapse pathway was the most significant biological pathway identified using those overlapping genes. GABA is the most common inhibitory neurotransmitter, and it plays a significant role in the development of addiction [[180]50]. We observed a significant positive correlation between plasma FGF21 and GABA concentrations in AUD patients. However, the effect size is small which might be due, at least in part, to heterogeneity of alcohol use disorder and biological variation among patients suffering from this disorder. In parallel, naïve FGF21 and PF-05231023 significantly induced GABA levels in iPSC-derived forebrain neurons and forebrain organoids. Opoku et al. reported that FGF21 regulates GABA concentrations in the brain, liver, and serum of mice with hepatic encephalopathy [[181]51]. However, it is not fully understood how FGF21 regulates GABA. We observed that a series of genes related to GABA catabolism were down-regulated in response to naïve FGF21 or PF-05231023 ([182]Figure 5A). We also demonstrated a drug-dependent occupancy of REST, EZH2, and H3K27me3 in the promoter regions of genes associated with GABA catabolism, which resulted in transcriptional repression in iPSC-derived forebrain neurons ([183]Figure 6). GABA regulation has been associated with histone monomethylation or trimethylation, and histone demethylation [[184]52]. Chromatin remodeling at GABAergic gene promoters, including GAD1, and GAD2 histone methylation, has been observed throughout an extended period of normal human pre-frontal cortex development, and plays a role in the neurobiology of schizophrenia [[185]53]. H3K27me3, a repressive chromatin marker, was present at these promoter sites [[186]53]. This is consistent with our findings ([187]Figure 6). Furthermore, our results demonstrate that FGF21 mediated the occupancy of REST, EZH2 and H3K27me3. As expected, we observed a negative correlation between mRNA expression of GABAergic gene promoters and the occupancy of the REST-EZH2-H3K37me3 complex in iPSC-derived forebrain neurons ([188]Figure 5, [189]Figure 6). These data, taken together, support our hypothesis that the drug-dependent occupancy of the REST-EZH2-H3K37me3 complex could explain the difference in gene expression of GABAergic genes in response to drug treatment. Collectively, our findings suggest that a previously uncharacterized epigenetic mechanism regulates GABAergic gene promoters by FGF21. These results also demonstrate that iPSC-derived brain-like cells could help to generate testable hypotheses for functional mechanistic studies, and, ultimately, add mechanistic rigor to future clinical trials of AUD therapeutics. We demonstrated that FGF21 increases GABA production by REST-EZH2-H3K27me3 mediated transcriptional repression of genes associated with GABA catabolism. GAD2 is responsible for catalyzing the production of GABA ([190]Figure 7). Our results showed that GABA concentrations were induced by FGF21, however, mRNA expression of GAD2 was repressed by FGF21. We also observed that ABAT, a crucial enzyme responsible for GABA catabolism, and SLC32A1 (an influx transporter for GABA), also displayed decreased expression in response to FGF21 treatment. As a result, we observed an increase of GABA levels within the culture medium of iPSC-derived neurons and brain organoids as a result of decreased degradation in the face of decreased biosynthesis. These results may suggest a potential negative feedback loop because of the induction of GABA by FGF21. Our study also has limitations. We began by performing RNA-seq using iPSC-derived forebrain neurons. Our sample size is small, which limits the generalizability of our findings. The human brain is complex, and one cell type in a specific brain region may not represent the complex human central nervous system. Future studies that include different brain regions will be required to pursue the results reported here, as well as the application of single-cell sequencing in iPSC-derived brain organoids in an attempt to identify cell-type specific, and drug-specific effects. In addition, further study is also warranted to explore the links between GABAergic chromatin remodeling and electrophysiological changes in response to naïve FGF21 and PF-05231023. Those results could provide additional functional insights into both of the compounds studied. Despite these limitations, our study provides novel mechanistic insights into the drug actions of naïve FGF21 and PF-05231023. 5. Conclusions In summary, this study demonstrates a novel mechanism of FGF21. Our results show that FGF21 can induce GABA concentrations in both iPSC-derived forebrain neurons and forebrain organoids. GABA is synthesized primarily from glutamate. GABA is then stored in synaptic vesicles via a vesicular GABA transporter (SLC32A1). GABA has complex regulatory functions for monoamine neurotransmission. GABA activity is terminated at synapses by uptake into both nerve terminals and surrounding cells via plasma member GABA transporters. GABA is then catabolized by GABA transaminase (ABAT) [[191]47]. Those genes related to GABA catabolism can be suppressed by naïve FGF21 or PF-05231023. This might be, at least in part, because FGF21 and PF-05231023 can facilitate the recruitment of REST-EZH2-H3K37me3 to GABAergic gene promoters, which, in turn, represses gene expression. Taken together, these results may provide novel mechanistic insights into FGF21 action in the CNS. Funding This work was supported in part by National Institutes of Health [grant numbers R01 AA27486, P20 AA17830, DA57928, and K01 AA28050]; Brain & Behavior Research Foundation, the Mayo Clinic Research Pipeline K2R Program, the Terrance and Bette Noble Foundation, and the Mayo Clinic Center for Individualized Medicine. The content of this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Author contributions MH wrote the first draft of the manuscript and supervised the study. MH designed the research; MH, CZ, and IM performed the research. MH, and CZ analyzed the data; CZ, BC. JB, and HL contributed analytical tools. TO, MS, PC, VK, QN, CS, and MS recruited study subjects, and provided administrative support. MH and RW obtained funding. All authors have given final approval of the version to be published. Acknowledgments