Shared and unique 3D genomic features of substance use disorders across multiple cell types =========================================================================================== * Khanh B. Trang * Alessandra Chesi * Sylvanus Toikumo * James A. Pippin * Matthew C. Pahl * Joan M. O’Brien * Laufey T. Amundadottir * Kevin M. Brown * Wenli Yang * Jaclyn Welles * Dominic Santoleri * Paul M. Titchenell * Patrick Seale * Babette S. Zemel * Yadav Wagley * Kurt D. Hankenson * Klaus H. Kaestner * Stewart A. Anderson * Matthew S. Kayser * Andrew D. Wells * Henry R. Kranzler * Rachel L. Kember * Struan F.A. Grant ## ABSTRACT Recent genome-wide association studies (GWAS) have revealed shared genetic components among alcohol, opioid, tobacco and cannabis use disorders. However, the extent of the underlying shared causal variants and effector genes, along with their cellular context, remain unclear. We leveraged our existing 3D genomic datasets comprising high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq and RNA-seq across >50 diverse human cell types to focus on genomic regions that coincide with GWAS loci. Using stratified LD regression, we determined the proportion of genome-wide SNP heritability attributable to the features assayed across our cell types by integrating recent GWAS summary statistics for the relevant traits: alcohol use disorder (AUD), tobacco use disorder (TUD), opioid use disorder (OUD) and cannabis use disorder (CanUD). Statistically significant enrichments (*P*<0.05) were observed in 14 specific cell types, with heritability reaching 9.2-fold for iPSC-derived cortical neurons and neural progenitors, confirming that they are crucial cell types for further functional exploration. Additionally, several pancreatic cell types, notably pancreatic beta cells, showed enrichment for TUD, with heritability enrichments up to 4.8-fold, suggesting genomic overlap with metabolic processes. Further investigation revealed significant positive genetic correlations between T2D with both TUD and CanUD (FDR<0.05) and a significant negative genetic correlation with AUD. Interestingly, after partitioning the heritability for each cell type’s cis-regulatory elements, the correlation between T2D and TUD for pancreatic beta cells was greater (r=0.2) than the global genetic correlation value. Our study provides new genomic insights into substance use disorders and implicates cell types where functional follow-up studies could reveal causal variant-gene mechanisms underpinning these disorders. ## INTRODUCTION Substance use disorders (SUDs) are a group of treatable psychiatric disorders that are associated with a variety of adverse outcomes. SUDs pose a pervasive global health challenge, affecting individuals across diverse demographic groups and populations. SUD severity varies from mild to severe and adverse outcomes generally correlate with the severity of the disorder. Alcohol, socially accepted and widely available, is one of the most commonly misused substances worldwide [1], leading to significant health, social, and economic losses. Excessive alcohol consumption is a major cause of premature death and disability, particularly among individuals aged 20-39, with about 13.5% of deaths in this group linked to alcohol. In the United States, 10.5% of individuals aged 12 and older suffer from alcohol use disorder (AUD), with over 178,000 annual alcohol-related deaths reported by the Centers for Disease Control and Prevention [2,3]. Tobacco use disorder (TUD), affecting 22.3% of the global population, is the most prevalent substance use disorder, causing nearly 8 million preventable deaths annually due to diseases such as cardiovascular and respiratory disorders, as well as various cancers [4]. The prevalence of opioid use disorder (OUD) has surged over the past three decades, driven by prescription opioid misuse and illicit opioids like fentanyl, resulting in approximately 81,806 US opioid overdose deaths in 2022 [5]. Cannabis use disorder (CanUD) has become more prominent with increasing legalization and is associated with morbidity, including certain cancers, cognitive impairments, and schizophrenia, contributing to societal problems that include reduced productivity and accidents [6–9]. SUDs have strong familial inheritance patterns [10], with heritability estimates from twin studies for these disorders of averaging approximately 50%. AUD heritability estimates range from 0.50 to 0.64 [11,12] and are higher for the diagnosis than traits like alcohol use initiation (0.30–0.40) [13] and frequency of consumption (0.37–0.47) [14], suggesting a greater genetic influence on the progression to problematic use. TUD heritability ranges from 0.30 to 0.70, depending on the assessment method used [15,16]. CanUD heritability ranges from 0.51 to 0.59, with environmental and genetic contributors reported across both use and misuse [17,18]. OUD heritability is around 50%, with 38% of variation attributed to opioid-specific genetic factors [19]. Studies also highlight that in addition to specific genetic influences for use disorders [20,21], there are broader heritable factors influencing SUD susceptibility in general [22]. Over the past ten years, there have been multiple large genome-wide association studies (GWAS) of SUDs, yielding a growing number of genome-wide significant loci [23,24]. Substantial progress has been made in identifying AUD loci, starting with the genes encoding the alcohol metabolic enzymes *ADH1B* and *ALDH2* [25–33]. Recently, larger samples have yielded additional loci, include *DRD2, GCKR, KLB* and *SLC39A8* [27,29,31,32,34,35]. While genetic factors play a significant role in shaping smoking behaviors, recent GWAS have primarily focused on identifying risk variants associated with nicotine consumption and TUD. These studies consistently link nicotine dependence to cholinergic nicotinic receptor genes, especially at *CHRNA5-CHRNA3-CHRNB4* locus [24], and a variant at the *DNMT3B* locus with heavy smoking and lung cancer risk. Recent multi-ancestral meta-analyses revealed 72 independent risk loci for TUD [36], and more than a thousand loci associated with various smoking phenotypes [37]. Due to smaller available samples for GWAS, fewer replicable loci have been uncovered for CanUD. The largest meta-GWAS of CanUD to date [38] identified several significant loci unique both to individual ancestries and in the multi-ancestry analysis, with key findings that include loci near *SLC36A2*, *SEMA6D*, *MCCC2*, *LRRC3B*, *PDE4B*, *LAMB2*, *FOXP1*, *GABRB1*, *MAGI2*, *SCAI*, *DRD2* and *ZFHX3* and confirming loci near *CHRNA2* [39] and *FOXP2* [40]. GWAS for OUD associations have revealed *CNIH3*, *KCNG2*, *APBB2*, *RGMA*, *KCNC1*, and *OPRM1* loci [34,41–43]. Furthermore, a large-scale GWAS integrating data from seven cohorts identified OUD variant associations at *OPRM1*, *FURIN*, and 18 other loci using multi-trait methods [44]. A second cross-ancestry meta-analysis uncovered 14 loci for OUD, 12 of which were novel, including *RABEPK*, *FBXW4*, *NCAM1* and *KCNN1* [45]. Despite this progress, we are still far from a mechanistic understanding of the effector genes and the cellular contexts operating at these genetic loci, as GWAS can only identify a genomic region associated with a trait, rather than a specific effector gene, and is cell-type agnostic. Tools like partitioned Linkage Disequilibrium Score Regression (LDSC) quantify the proportion of genome-wide SNP-heritability for a trait attributable to functional genomics categories of one’s choice, using information from all SNPs and explicitly modeling LD [46]. By combining 3D chromatin maps (Hi-C, Capture-C) with matched chromatin accessibility data (ATAC-seq), we sought to elucidate the specific cellular settings in which genetic variation contributes to the risk of AUD, TUD, OUD, and CanUD using partitioned LDSC. Capitalizing on existing GWAS results and our genomics datasets for >50 cell types, and leveraging LDSC, we report significant enrichments of the SUD GWAS signals in regulatory elements of specific cell types, shedding light on the etiology of susceptibility to SUDs. ## METHODS ### Data and resource **Table S1** lists the datasets used in prior studies. The original published studies provided configurations and technical details in ATAC-seq, Hi-C, and Capture-C ***library generation***. ### ATAC-seq preprocessing and peak calling Open chromatin regions (OCRs) were called using the ENCODE ATAC-seq pipeline ([https://github.com/ENCODE-DCC/atac-seq-pipeline](https://github.com/ENCODE-DCC/atac-seq-pipeline)). Reads were aligned to the GRCh37/hg19 or GRCh38/hg38 assembly genome using **bowtie2** [47], duplicates were removed, alignments from all replicates were pooled, and narrow peaks were called using **MACS2**. We lifted all coordinates from GRCh37/hg19 to GRCh38/hg38 to ensure consistency between datasets [48]. ### Promoter Capture-C pre-processing and interaction calling Paired-end reads were pre-processed using the **HICUP** pipeline [49] with **bowtie2** and GRCh37/hg19. Significant promoter interactions were called using unique read pairs using **CHICAGO** [50]. We analyzed individual fragments (1frag) and binned four fragments to improve long-distance sensitivity [51]. Interactions with CHICAGO score > 5 at either 1-fragment or 4-fragment resolution were considered significant. Interactions from both resolutions were merged and lifted from GRCh37/hg19 to GRCh38/hg38. ### Hi-C pre-processing and interaction calling As described in our recent study [52], paired-end reads from each replicate were pre-processed using the **HICUP v0.7.4** pipeline [49] and aligned by **bowtie2** with GRCh38/hg38. The alignment files were parsed and processed by **pairtools v0.3.0** [53] and indexed and compressed by **pairix v0.3.7** [54], then converted to Hi-C matrix binary format .*cool* at multiple resolutions (500 bp; 1, 2, 4, 10, 40, and 500 kbp; and 1 Mbp) by **cooler v0.8.11** [55] and normalized using the ICE method [56]. The matrices from different replicates were merged at each resolution using **cooler**. **Mustache v1.0.1** [57] and **Fit-Hi-C2 v2.0.7** [58] were used to call significant cis-interaction loops from merged replicate matrices at three resolutions—1 kb, 2 kb, and 4 kb—with the significance threshold at p-value < 0.1 and FDR < 1×10−6, respectively. The identified interaction loops were merged between the two tools, and then merged across resolutions prioritizing higher resolution for overlaps. ### Definition of cis-Regulatory Elements (cREs) We intersected ATAC-seq OCRs of each cell type with chromatin interaction loops determined by Hi-C/Capture-C of the same cell type, and with promoters (-1,500/+500 bp of TSS) defined by GENCODE v40. ### Reformatting of the GWAS summary statistics for the four substance use disorders **Table S2** lists the studies from which we drew European-population GWAS summary statistics for each of the four SUDs. We applied *--merge-alleles* with the list of HapMap3 variants to standardize all the GWAS summary statistics files. The baseline model LD scores, plink files, allele frequencies, HapMap3 variants list and regression weight files for the European 1000 genomes project phase 3 in GRCh38 were downloaded from [https://alkesgroup.broadinstitute.org/LDSCORE/GRCh38/](https://alkesgroup.broadinstitute.org/LDSCORE/GRCh38/). ### Cell type specific partitioned heritability of each trait We used **LDSC v.1.0.1** with *--h2* flag [46] to estimate the SNP-based heritability of each trait within 5 sets of input regions from each cell type: (1) OCRs, (2) OCRs at gene promoters, (3) cREs, (4) cREs with an expanded window of ±500 bp, (5) OCRs that were not cREs and not at a gene promoter. Each set of input regions from each cell type was used to create the annotation, which in turn was used to compute annotation-specific LD scores for each cell type region of interest. These annotation-specific LD scores were used with 53 categories of the full baseline model (v2.2) to compute partitioned heritability. ### Genetic correlation analysis We used **LDSC** with --rg flag [59] to compute the genetic correlations between each of the SUDs and type 2 diabetes (T2D) using European-ancestry meta-analysis summary statistics from the most recent T2D GWAS [60]. The genetic correlation between each pair of traits was computed with unconstrained intercepts. We computed global genetic correlations using standardized variants from the GWAS of each trait. We then partitioned the variants of each trait into cREs of each cell type and recomputed the cell type-specific genetic correlation between each SUD and T2D. ## RESULTS ### Enrichment patterns across varying open chromatin region definitions To investigate the enrichment of GWAS-discovered genetic variants associated with the SUDs across the different cell types, we employed Stratified Linkage Disequilibrium Score Regression (S-LDSR) [46] across all cis-regulatory elements (cREs) [61,62] identified through ATAC-seq and chromatin capture/Hi-C analysis for each cell type (**Figure 1)**. We integrated our repertoire of 59 cell types (**Table S1**) with the recent European-population-GWAS summary statistics for TUD, AUD, CanUD and OUD (**Table S2**). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/07/19/2024.07.18.24310649/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2024/07/19/2024.07.18.24310649/F1) Figure 1: Heritability enrichment of substance use disorders across diverse cell types 1st left column: Bar-plot shows total number of open chromatin regions (OCRs) identified by ATAC-seq for each cell type on bulk cells (green), or on single cell (pink); proportion of cREs identified by hi-C (yellow), or Capture-C (blue). 12 dot-plot panels display the heritability enrichment (LDSC analysis) for each cell type across 4 SUDs in 3 categories of OCRs: **Total OCRs**: all OCRs identified by ATAC-seq **Promoter OCRs**: OCRs overlapped with gene promoters **cREs**: of each cell type Whiskers represent enrichment standard errors. Grey-scaled dots correspond to P-values in -log10, with red asterisks indicating significant *P*-values (< 0.05). Dot size corresponds to the proportion of SNP contribution to heritability. Dashed line at 1 indicates no enrichment. We assessed cell-type specific enrichment of GWAS signals in three genomic regions: 1) **Total OCR**: Open chromatin regions defined by ATAC-seq; 2) **Promoter OCR**: OCRs overlapping a gene promoter; 3) **cREs**: OCRs not overlapping a promoter but showing a chromatin loop with a promoter (putative enhancers/suppressors). Different GWAS variants can affect a phenotype by regulating gene expression through mechanisms like altering enhancer function (cREs) or transcription factor binding at promoters (Promoter OCRs). As a control, we also assessed open chromatin regions not overlapping a cRE or promoter (“not-cREs/Prom OCRs”), expecting no significant enrichments. For all cell types, the variants within the total set of OCRs (**Fig.1 “Total OCRs” columns**) showed a positive risk heritability (Enrichment >1) for at least one disorder, including in 44 cell types for AUD, 51 for TUD, 53 for CanUD, and 31 for OUD. Of these, only 14 unique cell types (12 for TUD, 4 for AUD, 6 for CanUD and 2 for OUD) showed statistically significant enrichments (*P*<0.05). When limiting the analysis to OCRs at gene promoters (**Fig.1 “Promoter OCRs” columns**), the enrichments were significantly decreased for immune cell types, while being increased significantly for metabolic and other cell types. This OCR category also yielded the greatest variability of enrichment (Enrichment standard error) in all cell types and all disorders (**Fig S1**). Further constraining the LD enrichment assessment to include only OCRs that putatively regulate gene expression via chromatin contacts with gene promoters (**Figure 1 “cREs” column**) significantly increased the enrichment for most of the cell types for all disorders, decreased the *P*-values, and moderately narrowed the dispersion of enrichment ranges across different cell types compared to the promoter OCRs. The original S-LDSR method analyzed enrichment in the 500-bp flanking regions of their regulatory categories [46]. Our expanded analysis of the ±500 bp window for our cREs incorporated more weighted variants in the enrichment (**Fig S1 – Prop.SNP**), nearly as many as for the total OCRs in most instances. For GWAS signal distributions with sharp peaks, expanding the region by 500 bp dilutes the signal and increases P-values and enrichment standard errors without increasing heritability enrichment, as previously observed [63]. However, for SUDs, this expansion yielded stronger signals in LDSC statistics, lower P-values and enrichment standard errors, and greater enrichment across cell types. This suggests that the greater effect size of genetic signals for these disorders spans broader regions than our cREs. Conversely, when we analyzed OCRs located outside both cREs and promoters (“not-cREs/Prom OCRs”) as a control assessment (**Fig S2**), we observed generally lower enrichment than for the cREs with their expanded regions. However, an opposite pattern of enrichment was observed across the cell types (R = -0.01 to -0.2 comparing enrichment of “cREs” versus “not-cREs/Prom OCRs”, **Fig S3**), suggesting distinct functions of the GWAS variants attributed to the phenotype within different cellular system settings. (Detailed results in **Table S3**). Some cell types showed positive enrichment across all OCR categories, albeit at different levels of significance, though the numbers differed for the four disorders: 25 cell types for AUD, 29 for TUD, 6 for OUD, and 24 for CanUD. The iPSC-derived cortical neurons and pancreatic beta cells are two cell types that consistently showed positive enrichment across all OCR categories for all four disorders. ### Diverse enrichment patterns of SUDs across cell types and chromatin regions The general patterns mentioned above varied across the different SUDs. The AUD enrichment was more evident in the neural cell types (**Fig S4A**). iPSC-derived cortical neurons and neural progenitor cells and embryonic stem cell (ESC)-derived hypothalamic NPCs and neurons showed significantly positive enrichment across almost all OCR categories. Only astrocytes showed significant enrichment within “not-cREs/Prom OCRs”. On the other hand, several liver and pancreatic cell types consistently displayed positive enrichment in all OCR categories. Interestingly, hepatocytes and pancreatic beta cells showed significant enrichment for AUD within total OCRs and “not-cREs/Promoter OCRs”, while pancreatic alpha cells showed significant enrichment for the expanded cRE regions. TUD also revealed highly significant enrichment within neural cell types, with greater strength within the cREs and promoter OCR regions, and less enrichment within the control OCR categories (**Fig S4B**). Interestingly, the pancreatic alpha and beta cell types displayed significant enrichment consistently across all OCR region categories. Skeletal myotubes, melanocytes and hMSCs also showed enrichment for TUD loci within promoter OCRs and cRE types, while some naïve T-cell types displayed significant enrichment for the “not-cREs/Prom OCRs” regions. CanUD displayed a contrasting enrichment pattern for OCR categories (**Fig S4C**). The neural cell types showed enhanced enrichment with the ‘not promoter-OCRs or cREs’ regions. Metabolic cell types, including several pancreatic, liver, myotube, and osteoblast cell types, yielded significant enrichment for cRE regions. This disorder also showed the highest involvement of immune cell types, especially various T-helper cells. OUD signals had the most sporadic pattern and the greatest number of negative enrichments (**Fig S4D**). Nonetheless, we observed significant positive enrichments for cortical neurons and pancreatic beta cells. ### Cortical neurons, neural progenitors, and pancreatic cells as key players in SUD pathophysiology and therapeutic targets Despite the observed diversity in patterns across distinct SUDs, several cell types consistently exhibited significant levels of enrichment. Notably, iPSC-derived cortical neurons and neural progenitors, ESC-derived hypothalamus NPCs and neurons, and pancreatic alpha and beta cell types were notably consistent across the four disorders (**Fig S5**). The observed pattern of enrichment within cREs and their expanded regions suggest a pronounced association with the genetic architecture underlying SUDs. Conversely, there was a significantly lower level of enrichment within the contrasting OCR outside of both cREs and promoters (“not-cREs/Prom OCRs”). The involvement of pancreatic cell types in SUDs prompted us to consider the possibility of pleiotropy between SUDs and metabolic diseases such as type 2 diabetes. The moderate genome-wide positive genetic correlation (R=0.1) between T2D and both TUD and CanUD was significant, whereas it was significantly negative with AUD (**Fig S6**). Interestingly, after partitioning the heritability to each cell type’s cREs, the correlation between T2D and TUD within pancreatic beta cells was significantly higher than the genome-wide correlation value (R=0.2) (**Fig S6A**). The significance was established with a bootstrap t-test yielding a p-value of 7e-48 (**Fig S6B**). This implies that pancreatic beta cells are crucial in understanding the genetic overlap between T2D and TUD, possibly due to shared pathways or regulatory mechanisms active in these cells. This finding highlights the importance of examining cell-type-specific genetic contributions to understand more fully the complex relationship between metabolic disorders and SUDs. ## DISCUSSION We sought to identify cellular contexts mediating the genetic etiology of SUDs by examining enrichment patterns of epigenetic features across diverse cell types. Our approach integrated GWAS summary statistics with ATAC-seq, and promoter Capture C/Hi-C data. Utilizing LD score regression, quantifying polygenic effects and confounding factors, we quantified the contribution of SNP-based heritability to SUD pathogenesis. The observed positive heritability enrichment across various open chromatin features spanning multiple cell types revealed the involvement of metabolic and neural systems. Within our repertoire of cell types, we hypothesized that most of the GWAS signals would reside in cREs or gene promoters, as shown previously [61–68]. The enrichment patterns observed for AUD and TUD generally aligned with this. However, for CanUD and OUD, the GWAS signals exhibited a distinct pattern not fully captured by our cREs. This complexity makes it challenging to identify and characterize all relevant regulatory elements contributing to the susceptibility to these disorders. The presumed temporal nature of open chromatin regions within a given cell type likely also contributes to less observed enrichment within certain cell type cREs [69]. Notably, cortical neurons and pancreatic cells were consistently positively enriched across all OCR categories for all SUDs, suggesting their potentially crucial role in the pathophysiology of SUDs. The enrichment of cortical neurons and neural progenitor cells in SUDs aligns with accumulating evidence that implicates these cell types in the neurobiology of addiction [70–76]. The apparent involvement of pancreatic alpha and beta cells in SUDs represents a novel and intriguing insight into their pathology, underscoring the interplay between metabolic regulation and addictive behaviors. Chronic exposure to substances like alcohol, opioids, and cannabinoids can disrupt pancreatic function, leading to dysregulated glucose metabolism and increased risk of metabolic disorders like diabetes [77–79]. Chronic alcohol consumption increases risk of pancreatic diseases such as pancreatitis and pancreatic cancer, likely due to the toxic effects of alcohol on pancreatic tissue. However, genetic predisposition can influence the susceptibility of pancreatic tissue to alcohol-induced toxicity, making certain individuals more vulnerable to these diseases. Alcohol-induced pancreatic damage can impair insulin secretion, cause beta-cell dysfunction, and dysregulate glucagon secretion, contributing to metabolic abnormalities and the development of diabetes [80–82]. Opioid drugs exert direct effects on pancreatic alpha and beta cells. For example, opioids modulate insulin and glucagon secretion by pancreatic islet cells, potentially contributing to the glucose dysregulation and metabolic disturbances observed in opioid users [83,84]. Endocannabinoid receptors, including cannabinoid receptor 1 (CB1), are expressed in pancreatic alpha and beta cells, suggesting a direct influence of cannabinoids on pancreatic function. The activation of CB1 receptors can impair insulin secretion from beta cells and stimulate glucagon release from alpha cells, implicating the endocannabinoid system in the regulation of pancreatic hormone secretion and glucose metabolism [85–87]. Furthermore, a recent study uncovered a direct link between pancreatic cells and nicotine use, mediated by the key diabetes-associated transcription factor *TCF7L2* [88]. The study showed that TCF7L2 regulates nicotine intake by modulating nicotinic receptors in the habenula, a brain region involved in stress responses. This regulation influences blood glucose levels through signaling via the autonomic nervous system to the pancreas, with chronic nicotine use disrupting normal glucose regulation. Interestingly, pancreatic beta cells share features with the brain, including a common set of expressed genes, which suggests an evolutionary link [89]. During pancreatic organogenesis, the depression of Polycomb enables beta cells to present with a neuronal gene expression program [90], with the overall beta cells’ gene expression pattern and chromatin marks being closer to neuronal tissue types than other tissues. Insulin-producing neurons precede beta cells phylogenetically, with rodents, human (in vitro), and most invertebrates having neurons that produce insulin to manage blood glucose levels [91]. Given the shared features of pancreatic beta cells and neurons that extend from transcription to post-transcriptional regulation [92], understanding brain-pancreas interactions could aid in understanding the relationship between addiction and metabolic health. Several cell types exhibited a degree of negative heritability enrichment, with all traits yielding a negative enrichment in at least one cell type. Significant negative enrichments were observed in a few cell types, for different disorders, and for different OCR categories. However, these cell types displayed typical levels of enrichment for other traits and those traits demonstrated normal enrichment ranges across other cell types. This suggests that the observed negative enrichments are unlikely to stem from misclassified alleles in trait summaries or biased genetic regions of interest within cell types. Our findings in immune cell types are consistent with numerous studies that have shown the influence of SUDs on the immune response system and associated cell types. For instance, there are immunomodulatory effects of chronic alcohol consumption, including alterations in cytokine production, heightened CNS inflammation, impaired immune cell function, and increased susceptibility to infection [93–97]. Similarly, tobacco use has been linked to systemic inflammation, immune dysregulation, and heightened risk of autoimmune diseases [98–104]. Additionally, opioid misuse has been shown to disrupt immune homeostasis, leading to immunosuppression [83,105–109]. Finally, cannabis use has been associated with immunomodulatory effects, such as altered cytokine profiles, impaired T-cell function, and dysregulated immune cell signaling [110–114]. In considering these effects, it is important to factor in the effective sample sizes of the GWAS efforts, which can introduce noise and contribute to negative enrichments observed in regression analyses. This limitation, inherent to partial linkage regression methodologies, highlights the need for careful interpretation of disease variant enrichment in specific cellular contexts. In conclusion, by elucidating relevant regulatory regions across various cell types, we gained insight into the genetic underpinnings of SUDs. Our study has indicated in which specific cellular environments the genetic susceptibility for SUDs appear to lie. These observations warrant further research aimed at unraveling the underlying causes of these disorders in specific cellular contexts. This could offer new avenues for advancing our understanding of SUDs and aiding in the development of more effective targeted interventions. ## Supporting information Supplementary table S1 [[supplements/310649_file03.xlsx]](pending:yes) Supplementary table S2 [[supplements/310649_file04.xlsx]](pending:yes) Supplementary table S2 [[supplements/310649_file05.xlsx]](pending:yes) Supplementary figures [[supplements/310649_file06.pdf]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors ## AUTHOR CONTRIBUTIONS **Khanh B. Trang**: Conceptualization, Methodology, Formal analysis, Investigation, Data Curation, Writing - Original Draft, Visualization **Alessandra Chesi**: Formal analysis, Investigation, Data Curation, Writing-Reviewing and Editing **Sylvanus Toikumo**: Resources, Data Curation, Writing-Reviewing and Editing **James A. Pippin**: Resources **Matthew C. Pahl**: Methodology, Writing-Reviewing and Editing **Joan M. O’Brien**: Resources **Laufey T. Amundadottir**: Resources **Kevin M. Brown**: Resources **Wenli Yang**: Resources **Jaclyn Welles**: Resources **Dominic Santoleri**: Resources **Paul M. Titchenell**: Resources **Patrick Seale**: Resources **Babette S. Zemel**: Resources **Yadav Wagley**: Resources **Kurt D. Hankenson**: Resources **Klaus H. Kaestner**: Resources **Stewart A. Anderson**: Resources **Matthew S. Kayser**: Funding acquisition, Writing-Reviewing and Editing **Andrew D. Wells**: Resources, Writing-Reviewing and Editing **Henry R. Kranzler**: Supervision, Funding acquisition, Writing-Reviewing and Editing **Rachel L. Kember**: Supervision, Funding acquisition, Writing-Reviewing and Editing **Struan F.A. Grant**: Conceptualization, Supervision, Funding acquisition, Writing-Reviewing and Editing ## FUNDING This work was supported by NIAAA grant R01 AA030056 and the Mental Illness Research, Education and Clinical Center of the Crescenz VAMC. A.C. was supported by NHGRI grant R35 HG011959. S.F.A.G., W.Y., K.H.K and P.S were supported by NIDDK grant UM1 DK126194. S.F.A.G. and B.S.Z were supported by NICHD grant R01 HD100406. ## COMPETING INTERESTS Dr. Kranzler is a member of advisory boards for Dicerna Pharmaceuticals, Sophrosyne Pharmaceuticals, Enthion Pharmaceuticals, and Clearmind Medicine; a consultant to Sobrera Pharmaceuticals and Altimmune; the recipient of research funding and medication supplies for an investigator-initiated study from Alkermes; a member of the American Society of Clinical Psychopharmacology’s Alcohol Clinical Trials Initiative, which was supported in the last three years by Alkermes, Dicerna, Ethypharm, Lundbeck, Mitsubishi, Otsuka, and Pear Therapeutics; and a holder of U.S. patent 10,900,082 titled: “Genotype-guided dosing of opioid agonists,” issued 26 January 2021. The remaining authors have nothing to disclose. ## ACKNOWLEDGEMENTS S.F.A.G. is the Daniel B. Burke Endowed Chair for Diabetes Research. * Received July 18, 2024. * Revision received July 18, 2024. * Accepted July 19, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. WHO. Global status report on alcohol and health 2018. 2018. 2. SAMHSA. National Survey on Drug Use and Health. 2022:Table 2.25—Alcohol use in lifetime: among people aged 12 or older; by age group and demographic characteristics, 2021 and 22. 3. Esser MB SA, Liu Y, Naimi TS. Vol. 73 154–61 (MMWR and Morbidity and Mortality Weekly Report). 4. WHO. WHO global report on trends in prevalence of tobacco use 2000-2025. 2021. 5. Ahmad FB CJ, Rossen LM, Sutton P. (National Vital Statistics System, National Center for Health Statistics, 2021). 6. Hasin DS, Saha TD, Kerridge BT, Goldstein RB, Chou SP, Zhang H, et al. Prevalence of Marijuana Use Disorders in the United States Between 2001-2002 and 2012-2013. JAMA Psychiatry. 2015;72(12):1235–42. 7. WHO. The health and social effects of nonmedical cannabis use. 2016. 8. Lopez-Quintero C, Pérez de los Cobos J, Hasin DS, Okuda M, Wang S, Grant BF, et al. Probability and predictors of transition from first use to dependence on nicotine, alcohol, cannabis, and cocaine: results of the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC). Drug Alcohol Depend. 2011;115(1-2):120–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.drugalcdep.2010.11.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21145178&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291079400019&link_type=ISI) 9. Winters KC, Lee CY. Likelihood of developing an alcohol and cannabis use disorder during youth: association with recent use and age. Drug Alcohol Depend. 2008;92(1-3):239–47. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.drugalcdep.2007.08.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17888588&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000252707200030&link_type=ISI) 10. Prom-Wormley EC, Ebejer J, Dick DM, Bowers MS. The genetic epidemiology of substance use disorder: A review. Drug Alcohol Depend. 2017;180:241–59. 11. Heath AC, Bucholz KK, Madden PA, Dinwiddie SH, Slutske WS, Bierut LJ, et al. Genetic and environmental contributions to alcohol dependence risk in a national twin sample: consistency of findings in women and men. Psychol Med. 1997;27(6):1381–96. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1017/S0033291797005643&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9403910&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997YJ15400016&link_type=ISI) 12. Kendler KS. Twin studies of psychiatric illness: an update. Arch Gen Psychiatry. 2001;58(11):1005–14. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.58.11.1005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11695946&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000172056100002&link_type=ISI) 13. Koopmans JR, Slutske WS, van Baal GC, Boomsma DI. The influence of religion on alcohol use initiation: evidence for genotype X environment interaction. Behav Genet. 1999;29(6):445–53. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1023/A:1021679005623&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10857249&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000087465200007&link_type=ISI) 14. Viken RJ, Kaprio J, Koskenvuo M, Rose RJ. Longitudinal analyses of the determinants of drinking and of drinking to intoxication in adolescent twins. Behav Genet. 1999;29(6):455–61. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1023/A:1021631122461&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10857250&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000087465200008&link_type=ISI) 15. Sullivan PF, Kendler KS. The genetic epidemiology of smoking. Nicotine Tob Res. 1999;1 Suppl 2:S51–7; discussion S69-70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/14622299050011811&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11768187&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 16. Agrawal A, Verweij KJ, Gillespie NA, Heath AC, Lessov-Schlaggar CN, Martin NG, et al. The genetics of addiction-a translational perspective. Transl Psychiatry. 2012;2(7):e140. 17. Agrawal A, Lynskey MT. The genetic epidemiology of cannabis use, abuse and dependence. Addiction. 2006;101(6):801–12. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1360-0443.2006.01399.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16696624&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000237516100011&link_type=ISI) 18. Verweij KJ, Zietsch BP, Lynskey MT, Medland SE, Neale MC, Martin NG, et al. Genetic and environmental influences on cannabis use initiation and problematic use: a meta-analysis of twin studies. Addiction. 2010;105(3):417–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1360-0443.2009.02831.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20402985&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 19. Deak JD, Johnson EC. Genetics of substance use disorders: a review. Psychological Medicine. 2021;51(13):2189–200. 20. Tsuang MT, Lyons MJ, Meyer JM, Doyle T, Eisen SA, Goldberg J, et al. Co-occurrence of abuse of different drugs in men: the role of drug-specific and shared vulnerabilities. Arch Gen Psychiatry. 1998;55(11):967–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.55.11.967&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9819064&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000076748200002&link_type=ISI) 21. Kendler KS, Myers J, Prescott CA. Specificity of genetic and environmental risk factors for symptoms of cannabis, cocaine, alcohol, caffeine, and nicotine dependence. Arch Gen Psychiatry. 2007;64(11):1313–20. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.64.11.1313&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17984400&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250701700011&link_type=ISI) 22. Kendler KS, Prescott CA, Myers J, Neale MC. The structure of genetic and environmental risk factors for common psychiatric and substance use disorders in men and women. Arch Gen Psychiatry. 2003;60(9):929–37. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.60.9.929&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12963675&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185155800010&link_type=ISI) 23. Johnson EC, Chang Y, Agrawal A. An Update on the Role of Common Genetic Variation Underlying Substance Use Disorders. Current Genetic Medicine Reports. 2020;8(2):35–46. 24. Hancock DB, Markunas CA, Bierut LJ, Johnson EO. Human Genetics of Addiction: New Insights and Future Directions. Current Psychiatry Reports. 2018;20(2):8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11920-018-0873-3&link_type=DOI) 25. Edenberg HJ, McClintick JN. Alcohol Dehydrogenases, Aldehyde Dehydrogenases, and Alcohol Use Disorders: A Critical Review. Alcohol Clin Exp Res. 2018;42(12):2281–97. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/acer.13904&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30320893&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 26. Gelernter J, Kranzler HR, Sherva R, Almasy L, Koesterer R, Smith AH, et al. Genome-wide association study of alcohol dependence:significant findings in African- and European-Americans including novel risk loci. Molecular Psychiatry. 2014;19(1):41–49. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/mp.2013.145&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24166409&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000328964700011&link_type=ISI) 27. Kranzler HR, Zhou H, Kember RL, Vickers Smith R, Justice AC, Damrauer S, et al. Genome-wide association study of alcohol consumption and use disorder in 274,424 individuals from multiple populations. Nature Communications. 2019;10(1):1499. 28. Walters RK, Polimanti R, Johnson EC, McClintick JN, Adams MJ, Adkins AE, et al. Transancestral GWAS of alcohol dependence reveals common genetic underpinnings with psychiatric disorders. Nat Neurosci. 2018;21(12):1656–69. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41593-018-0275-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30482948&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 29. Clarke TK, Adams MJ, Davies G, Howard DM, Hall LS, Padmanabhan S, et al. Genome-wide association study of alcohol consumption and genetic overlap with other health-related traits in UK Biobank (N=112 117). Molecular Psychiatry. 2017;22(10):1376–84. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/mp.2017.153&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28937693&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 30. Gelernter J, Sun N, Polimanti R, Pietrzak RH, Levey DF, Lu Q, et al. Genome-wide Association Study of Maximum Habitual Alcohol Intake in >140,000 U.S. European and African American Veterans Yields Novel Risk Loci. Biological Psychiatry. 2019;86(5):365–76. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2019.03.984&link_type=DOI) 31. Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, et al. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nature Genetics. 2019;51(2):237–44. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0307-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 32. Sanchez-Roige S, Fontanillas P, Elson SL, Gray JC, de Wit H, Davis LK, et al. Genome-wide association study of alcohol use disorder identification test (AUDIT) scores in 20 328 research participants of European ancestry. Addict Biol. 2019;24(1):121–31. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/adb.12574&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29058377&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 33. Xu K, Kranzler HR, Sherva R, Sartor CE, Almasy L, Koesterer R, et al. Genomewide Association Study for Maximum Number of Alcoholic Drinks in European Americans and African Americans. Alcohol Clin Exp Res. 2015;39(7):1137–47. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/acer.12751&link_type=DOI) 34. Zhou H, Rentsch CT, Cheng Z, Kember RL, Nunez YZ, Sherva RM, et al. Association of OPRM1 Functional Coding Variant With Opioid Use Disorder: A Genome-Wide Association Study. JAMA Psychiatry. 2020;77(10):1072–80. 35. Sanchez-Roige S, Palmer AA, Fontanillas P, Elson SL, Adams MJ, Howard DM, et al. Genome-Wide Association Study Meta-Analysis of the Alcohol Use Disorders Identification Test (AUDIT) in Two Population-Based Cohorts. Am J Psychiatry. 2019;176(2):107–18. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2018.18040369&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30336701&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 36. Toikumo S, Jennings MV, Pham BK, Lee H, Mallard TT, Bianchi SB, et al. Multi-ancestry meta-analysis of tobacco use disorder identifies 461 potential risk genes and reveals associations with multiple health outcomes. Nat Hum Behav. 2024. 37. Saunders GRB, Wang X, Chen F, Jang S-K, Liu M, Wang C, et al. Genetic diversity fuels gene discovery for tobacco and alcohol use. Nature. 2022;612(7941):720–24. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-022-05477-4&link_type=DOI) 38. Levey DF, Galimberti M, Deak JD, Wendt FR, Bhattacharya A, Koller D, et al. Multi-ancestry genome-wide association study of cannabis use disorder yields insight into disease biology and public health implications. Nat Genet. 2023;55(12):2094–103. 39. Demontis D, Rajagopal VM, Thorgeirsson TE, Als TD, Grove J, Leppälä K, et al. Genome-wide association study implicates CHRNA2 in cannabis use disorder. Nat Neurosci. 2019;22(7):1066–74. 40. Johnson EC, Demontis D, Thorgeirsson TE, Walters RK, Polimanti R, Hatoum AS, et al. A large-scale genome-wide association study meta-analysis of cannabis use disorder. The Lancet Psychiatry. 2020;7(12):1032–45. 41. Cheng Z, Zhou H, Sherva R, Farrer LA, Kranzler HR, Gelernter J. Genome-wide Association Study Identifies a Regulatory Variant of RGMA Associated With Opioid Dependence in European Americans. Biol Psychiatry. 2018;84(10):762–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2017.12.016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 42. Gelernter J, Kranzler HR, Sherva R, Koesterer R, Almasy L, Zhao H, et al. Genome-Wide Association Study of Opioid Dependence: Multiple Associations Mapped to Calcium and Potassium Pathways. Biological Psychiatry. 2014;76(1):66–74. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2013.08.034&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24143882&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 43. Polimanti R, Walters RK, Johnson EC, McClintick JN, Adkins AE, Adkins DE, et al. Leveraging genome-wide data to investigate differences between opioid use vs. opioid dependence in 41,176 individuals from the Psychiatric Genomics Consortium. Mol Psychiatry. 2020;25(8):1673–87. 44. Deak JD, Zhou H, Galimberti M, Levey DF, Wendt FR, Sanchez-Roige S, et al. Genome-wide association study in individuals of European and African ancestry and multi-trait analysis of opioid use disorder identifies 19 independent genome-wide significant risk loci. Molecular Psychiatry. 2022;27(10):3970–79. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41380-022-01709-1&link_type=DOI) 45. Kember RL, Vickers-Smith R, Xu H, Toikumo S, Niarchou M, Zhou H, et al. Cross-ancestry meta-analysis of opioid use disorder uncovers novel loci with predominant effects in brain regions associated with addiction. Nature Neuroscience. 2022;25(10):1279–87. 46. Finucane HK, Bulik-Sullivan B, Gusev A, Trynka G, Reshef Y, Loh P-R, et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nature Genetics. 2015;47(11):1228–35. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3404&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26414678&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 47. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature Methods. 2012;9(4):357–59. 48. Hinrichs AS, Karolchik D, Baertsch R, Barber GP, Bejerano G, Clawson H, et al. The UCSC Genome Browser Database: update 2006. Nucleic Acids Research. 2006;34(suppl_1):D590–D98. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkj144&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16381938&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000239307700126&link_type=ISI) 49. Wingett S, Ewels P, Furlan-Magaril M, Nagano T, Schoenfelder S, Fraser P, et al. HiCUP: pipeline for mapping and processing Hi-C data. F1000Res. 2015;4:1310. 50. Cairns J, Freire-Pritchett P, Wingett SW, Várnai C, Dimond A, Plagnol V, et al. CHiCAGO: robust detection of DNA looping interactions in Capture Hi-C data. Genome Biology. 2016;17(1):1–17. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-015-0866-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26753840&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 51. C S, MC P, SFA G, AD W. Restriction enzyme selection dictates detection range sensitivity in chromatin conformation capture-based variant-to-gene mapping approaches. Human genetics. 2021;140(10). 52. Su C, Gao L, May CL, Pippin JA, Boehm K, Lee M, et al. 3D chromatin maps of the human pancreas reveal lineage-specific regulatory architecture of T2D risk. Cell Metabolism. 2022;34(9). 53. Open2C, Abdennur N, Fudenberg G, Flyamer IM, Galitsyna AA, Goloborodko A, et al. Pairtools: From sequencing data to chromosome contacts. PLOS Computational Biology. 2024;20(5):e1012164. 54. S L. A tool for indexing and querying on a block-compressed text file containing pairs of genomic coordinates. 2020. [https://github.com/4dn-dcic/pairix](https://github.com/4dn-dcic/pairix). 55. Abdennur N, Goloborodko A, Imakaev M, Kerpedjiev P, Fudenberg G, Oullette S, et al. 56. Imakaev M, Fudenberg G, McCord RP, Naumova N, Goloborodko A, Lajoie BR, et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nature Methods. 2012;9(10):999–1003. 57. Roayaei Ardakany A, Gezer HT, Lonardi S, Ay F. Mustache: multi-scale detection of chromatin loops from Hi-C and Micro-C maps using scale-space representation. Genome Biology. 2020;21(1):1–17. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-020-1935-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32033565&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 58. A K, S B, F A. Identifying statistically significant chromatin contacts from Hi-C data with FitHiC2. Nature protocols. 2020;15(3). 59. Bulik-Sullivan B, Finucane HK, Anttila V, Gusev A, Day FR, Loh P-R, et al. An atlas of genetic correlations across human diseases and traits. Nature Genetics. 2015;47(11):1236–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3406&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26414676&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 60. Suzuki K, Hatzikotoulas K, Southam L, Taylor HJ, Yin X, Lorenz KM, et al. Genetic drivers of heterogeneity in type 2 diabetes pathophysiology. Nature. 2024;627(8003):347–57. 61. Pahl MC, Doege CA, Hodge KM, Littleton SH, Leonard ME, Lu S, et al. Cis-regulatory architecture of human ESC-derived hypothalamic neuron differentiation aids in variant-to-gene mapping of relevant complex traits. Nature Communications. 2021;12(1):1–12. 62. Chesi A, Wagley Y, Johnson ME, Manduchi E, Su C, Lu S, et al. Genome-scale Capture C promoter interactions implicate effector genes at GWAS loci for bone mineral density. Nature Communications. 2019;10(1):1–11. 63. Trang KB, Pahl MC, Pippin JA, Su C, Littleton SH, Sharma P, et al. 3D genomic features across >50 diverse cell types reveal insights into the genomic architecture of childhood obesity. medRxiv. 2024. 64. Littleton SH, Trang KB, Volpe CM, Cook K, DeBruyne N, Maguire JA, et al. Variant-to-function analysis of the childhood obesity chr12q13 locus implicates rs7132908 as a causal variant within the 3’ UTR of FAIM2. Cell Genom. 2024;4(5):100556. 65. Su C, Argenziano M, Lu S, Pippin JA, Pahl MC, Leonard ME, et al. 3D promoter architecture re-organization during iPSC-derived neuronal cell differentiation implicates target genes for neurodevelopmental disorders. Progress in Neurobiology. 2021;201:102000. 66. Lasconi C, Pahl MC, Pippin JA, Su C, Johnson ME, Chesi A, et al. Variant-to-gene-mapping analyses reveal a role for pancreatic islet cells in conferring genetic susceptibility to sleep-related traits. Sleep. 2022;45(8). 67. Su C, Johnson ME, Torres A, Thomas RM, Manduchi E, Sharma P, et al. Mapping effector genes at lupus GWAS loci using promoter Capture-C in follicular helper T cells. Nature Communications. 2020;11(1):3294. 68. Conery M, Pippin JA, Wagley Y, Trang K, Pahl MC, Villani DA, et al. GWAS-informed data integration and non-coding CRISPRi screen illuminate genetic etiology of bone mineral density. bioRxiv. 2024. 69. de la Torre-Ubieta L, Stein JL, Won H, Opland CK, Liang D, Lu D, et al. The Dynamic Landscape of Open Chromatin during Human Cortical Neurogenesis. Cell. 2018;172(1):289–304.e18. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2017.12.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29307494&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 70. Koob GF, Volkow ND. Neurocircuitry of Addiction. Neuropsychopharmacology. 2010;35(1):217–38. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npp.2009.110&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19710631&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000272784600012&link_type=ISI) 71. Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry. 2016;3(8):760–73. 72. Koob GF. Neurobiology of Opioid Addiction: Opponent Process, Hyperkatifeia, and Negative Reinforcement. Biol Psychiatry. 2020;87(1):44–53. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.BIOPSYCH.2019.05.023&link_type=DOI) 73. Xu C, Loh HH, Law PY. Effects of addictive drugs on adult neural stem/progenitor cells. Cell Mol Life Sci. 2016;73(2):327–48. 74. Robinson TE, Kolb B. Structural plasticity associated with exposure to drugs of abuse. Neuropharmacology. 2004;47 Suppl 1:33–46. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuropharm.2004.06.025&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15464124&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000224797200005&link_type=ISI) 75. Montoya-Filardi A, Mazón M. The addicted brain: imaging neurological complications of recreational drug abuse. Radiologia. 2017;59(1):17–30. 76. Bhattacherjee A, Djekidel MN, Chen R, Chen W, Tuesta LM, Zhang Y. Cell type-specific transcriptional programs in mouse prefrontal cortex during adolescence and addiction. Nature Communications. 2019;10(1):4169. 77. Ojo O, Wang XH, Ojo OO, Ibe J. The Effects of Substance Abuse on Blood Glucose Parameters in Patients with Diabetes: A Systematic Review and Meta-Analysis. Int J Environ Res Public Health. 2018;15(12). 78. Bruggeman BS, Campbell-Thompson M, Filipp SL, Gurka MJ, Atkinson MA, Schatz DA, et al. Substance Use Affects Type 1 Diabetes Pancreas Pathology: Implications for Future Studies. Front Endocrinol (Lausanne). 2021;12:778912. 79. Reece AS, Hulse GK. Sociodemographically Stratified Exploration of Pancreatic Cancer Incidence in Younger US Patients: Implication of Cannabis Exposure as a Risk Factor. Gastroenterology Insights. 2023;14(2):204–35. 80. Kim JY, Lee DY, Lee YJ, Park KJ, Kim KH, Kim JW, et al. Chronic alcohol consumption potentiates the development of diabetes through pancreatic β-cell dysfunction. World J Biol Chem. 2015;6(1):1–15. 81. Steiner JL, Crowell KT, Lang CH. Impact of Alcohol on Glycemic Control and Insulin Action. Biomolecules. 2015;5(4):2223–46. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/biom5042223&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 82. Nelson NG, Weingarten MJ, Law WX, Sangiamo DT, Liang N-C. Joint and separate exposure to alcohol and Δ9-tetrahydrocannabinol produced distinct effects on glucose and insulin homeostasis in male rats. Scientific Reports. 2019;9(1):12025. 83. Moustafa SR. The immune-opioid axis in prediabetes: predicting prediabetes with insulin resistance by plasma interleukin-10 and endomorphin-2 to kappa-opioid receptors ratio. Diabetology & Metabolic Syndrome. 2021;13(1):61. 84. Koekkoek LL, van der Gun LL, Serlie MJ, la Fleur SE. The Clash of Two Epidemics: the Relationship Between Opioids and Glucose Metabolism. Current Diabetes Reports. 2022;22(7):301–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11892-022-01473-0&link_type=DOI) 85. Song D, Geetha HS, Jain S, Reyes JV, Jaiswal V, Nepal G, et al. Delayed presentation of cannabis induced pancreatitis. Clin Case Rep. 2022;10(3):e05595. 86. Qi M, Kaddis JS, Chen K-T, Rawson J, Omori K, Chen ZB, et al. Chronic marijuana usage by human pancreas donors is associated with impaired islet function. PLOS ONE. 2021;16(10):e0258434. 87. Cortes-Justo E, Garfias-Ramírez SH, Vilches-Flores A. The function of the endocannabinoid system in the pancreatic islet and its implications on metabolic syndrome and diabetes. Islets. 2023;15(1):1–11. 88. Duncan A, Heyer MP, Ishikawa M, Caligiuri SPB, Liu X-a, Chen Z, et al. Habenular TCF7L2 links nicotine addiction to diabetes. Nature. 2019;574(7778):372–77. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-019-1653-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31619789&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 89. Arntfield ME, van der Kooy D. β-Cell evolution: How the pancreas borrowed from the brain: The shared toolbox of genes expressed by neural and pancreatic endocrine cells may reflect their evolutionary relationship. BioEssays : news and reviews in molecular, cellular and developmental biology. 2011;33(8):582–7. 90. van Arensbergen J, García-Hurtado J, Moran I, Maestro MA, Xu X, Van de Casteele M, et al. Derepression of Polycomb targets during pancreatic organogenesis allows insulin-producing beta-cells to adopt a neural gene activity program. Genome Res. 2010;20(6):722–32. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjg6IjIwLzYvNzIyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDcvMTkvMjAyNC4wNy4xOC4yNDMxMDY0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 91. Devaskar SU, Giddings SJ, Rajakumar PA, Carnaghi LR, Menon RK, Zahm DS. Insulin gene expression and insulin synthesis in mammalian neuronal cells. J Biol Chem. 1994;269(11):8445–54. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjExOiIyNjkvMTEvODQ0NSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA3LzE5LzIwMjQuMDcuMTguMjQzMTA2NDkuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 92. Juan-Mateu J, Rech TH, Villate O, Lizarraga-Mollinedo E, Wendt A, Turatsinze JV, et al. Neuron-enriched RNA-binding Proteins Regulate Pancreatic Beta Cell Function and Survival. J Biol Chem. 2017;292(8):3466–80. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEwOiIyOTIvOC8zNDY2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDcvMTkvMjAyNC4wNy4xOC4yNDMxMDY0OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 93. Meredith LR, Burnette EM, Grodin EN, Irwin MR, Ray LA. Immune treatments for alcohol use disorder: A translational framework. Brain, Behavior, and Immunity. 2021;97:349–64. 94. Crews FT, Vetreno RP. Mechanisms of neuroimmune gene induction in alcoholism. Psychopharmacology. 2016;233(9):1543–57. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00213-015-3906-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25787746&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 95. Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacology Biochemistry and Behavior. 2019;177:34–60. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pbb.2018.12.007&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30590091&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 96. Osterndorff-Kahanek EA, Becker HC, Lopez MF, Farris SP, Tiwari GR, Nunez YO, et al. Chronic Ethanol Exposure Produces Time- and Brain Region-Dependent Changes in Gene Coexpression Networks. PLOS ONE. 2015;10(3):e0121522. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0121522&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25803291&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 97. Fan X, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Freedman ND, et al. Drinking alcohol is associated with variation in the human oral microbiome in a large study of American adults. Microbiome. 2018;6(1):59. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s40168-018-0448-x&link_type=DOI) 98. Elisia I, Lam V, Cho B, Hay M, Li MY, Yeung M, et al. The effect of smoking on chronic inflammation, immune function and blood cell composition. Scientific Reports. 2020;10(1):19480. 99. Hosseinzadeh A, Thompson PR, Segal BH, Urban CF. Nicotine induces neutrophil extracellular traps. Journal of Leukocyte Biology. 2016;100(5):1105–12. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1189/jlb.3AB0815-379RR&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27312847&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 100.Tran QT, Arimilli S, Scott E, Chen P, Prasad GL. Differences in biomarkers of inflammation and immune responses in chronic smokers and moist snuff users. Cytokine. 2021;137:155299. 101.Darabseh MZ, Maden-Wilkinson TM, Welbourne G, Wüst RCI, Ahmed N, Aushah H, et al. Fourteen days of smoking cessation improves muscle fatigue resistance and reverses markers of systemic inflammation. Scientific Reports. 2021;11(1):12286. 102.Saint-André V, Charbit B, Biton A, Rouilly V, Possémé C, Bertrand A, et al. Smoking changes adaptive immunity with persistent effects. Nature. 2024;626(8000):827–35. 103.Kridin K, Zamir H, Cohen AD. Cigarette smoking associates inversely with a cluster of two autoimmune diseases: ulcerative colitis and pemphigus. Immunologic Research. 2018;66(4):555–56. 104.Gomes JP, Watad A, Shoenfeld Y. Nicotine and autoimmunity: The lotus’ flower in tobacco. Pharmacological Research. 2018;128:101–09. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.phrs.2017.10.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29051105&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F07%2F19%2F2024.07.18.24310649.atom) 105.Kosciuczuk U, Knapp P, Lotowska-Cwiklewska AM. Opioid-induced immunosuppression and carcinogenesis promotion theories create the newest trend in acute and chronic pain pharmacotherapy. Clinics (Sao Paulo). 2020;75:e1554. 106.Kelty E, Rae K, Jantzie LL, Wyrwoll CS, Preen DB. Prenatal Opioid Exposure and Immune-Related Conditions in Children. JAMA Network Open. 2024;7(1):e2351933–e33. 107.Abu Y, Vitari N, Yan Y, Roy S. Opioids and Sepsis: Elucidating the Role of the Microbiome and microRNA-146. International Journal of Molecular Sciences. 2022;23(3):1097. 108.Trunfio M, Chaillon A, Beliakova-Bethell N, Deiss R, Letendre SL, Riggs PK, et al. Beyond the Syndemic of Opioid Use Disorders and HIV: The Impact of Opioids on Viral Reservoirs. Viruses. 2023;15(8). 109.O’Sullivan SJ, Malahias E, Park J, Srivastava A, Reyes BAS, Gorky J, et al. Single-Cell Glia and Neuron Gene Expression in the Central Amygdala in Opioid Withdrawal Suggests Inflammation With Correlated Gut Dysbiosis. Frontiers in Neuroscience. 2019;13. 110.Henriquez JE, Bach AP, Matos-Fernandez KM, Crawford RB, Kaminski NE. Δ(9)-Tetrahydrocannabinol (THC) Impairs CD8(+) T Cell-Mediated Activation of Astrocytes. J Neuroimmune Pharmacol. 2020;15(4):863–74. 111.Xiong X, Chen S, Shen J, You H, Yang H, Yan C, et al. Cannabis suppresses antitumor immunity by inhibiting JAK/STAT signaling in T cells through CNR2. Signal Transduction and Targeted Therapy. 2022;7(1):99. 112.Manuzak JA, Gott TM, Kirkwood JS, Coronado E, Hensley-McBain T, Miller C, et al. Heavy Cannabis Use Associated With Reduction in Activated and Inflammatory Immune Cell Frequencies in Antiretroviral Therapy-Treated Human Immunodeficiency Virus-Infected Individuals. Clin Infect Dis. 2018;66(12):1872–82. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/cix1116&link_type=DOI) 113.Nichols JM, Kaplan BLF. Immune Responses Regulated by Cannabidiol. Cannabis Cannabinoid Res. 2020;5(1):12–31. 114.Martini S, Gemma A, Ferrari M, Cosentino M, Marino F. Effects of Cannabidiol on Innate Immunity: Experimental Evidence and Clinical Relevance. International Journal of Molecular Sciences. 2023;24(4):3125.