Immunological Drivers and Potential Novel Drug Targets for Major Psychiatric, Neurodevelopmental, and Neurodegenerative Conditions ================================================================================================================================== * Christina Dardani * Jamie W. Robinson * Hannah J. Jones * Dheeraj Rai * Evie Stergiakouli * Jakob Grove * Renee Gardner * Andrew M. McIntosh * Alexandra Havdahl * Gibran Hemani * George Davey Smith * Tom G. Richardson * Tom R. Gaunt * Golam M. Khandaker ## Abstract Immune dysfunction is implicated in the aetiology of psychiatric, neurodevelopmental, and neurodegenerative conditions, but the issue of causality remains unclear impeding attempts to develop new interventions. We have tested evidence for causality for 735 immune response-related biomarkers on 7 neuropsychiatric conditions, using cutting-edge genomic causal inference methods (Mendelian randomization and genetic colocalization) applied to genomic data on protein and gene expression across blood and brain. We provide robust evidence of causality for 21 biomarkers, including two previously unreported (*LATS1*, and *FCN1*), confirming a role of both brain specific and systemic immune response in the pathogenesis of several neuropsychiatric conditions especially schizophrenia, Alzheimer’s disease, depression, and bipolar disorder. Furthermore, 18 of the identified biomarkers are therapeutically tractable, including *ACE*, *TNFRSF17*, and *CD40,* with drugs approved or in advanced clinical trials, offering an opportunity for repurposing existing drugs for neuropsychiatric indications. ## Introduction Psychiatric, neurodevelopmental, and neurodegenerative conditions (henceforth, neuropsychiatric conditions) are among the leading causes of disability worldwide1,2. These conditions are typically chronic, and affect mood, perception, cognition, and behaviour. Biological pathways contributing to these conditions are poorly understood, impeding attempts to identify effective new interventions3. For example, approximately one in three individuals with depression or schizophrenia do not respond to current medications which primarily target monoamine neurotransmitters4. This suggests that the current one-size-fits-all approach to treatment for these conditions may not be tenable. Therefore, identifying biological pathways underpinning neuropsychiatric conditions to help prioritise novel intervention targets remains a key priority for mental health research5. Over the last two decades immune dysfunction has emerged as a promising mechanistic candidate for several neuropsychiatric conditions. For example, immune activating drugs induce depressive symptoms in hepatitis C patients6 and healthy volunteers7. Meta-analyses of case-control studies confirm atypical levels of cytokines in blood plasma and cerebrospinal fluid of individuals with schizophrenia, depression, and bipolar disorder8,9. Neuroimaging with positron emission tomography shows evidence of neuroinflammation in acute depression10. Nationwide cohort studies indicate associations between autoimmune conditions, infections and neuropsychiatric conditions, such as schizophrenia11,12, attention deficit hyperactivity disorder (ADHD)13, Alzheimer’s disease14, and depression15. However, inferring causality remains an important outstanding issue because the observed associations could be result of residual confounding or reverse causation. Mendelian randomisation (MR), a genetic causal inference method that can minimise these limitations by using genetic variants regulating levels/activity of biomarkers as proxies16–18, has provided some evidence for a potential causal effect of IL-6 and CRP in depression and schizophrenia19,20. RCTs suggest that broad spectrum anti-inflammatory drugs improve mood and psychotic symptoms in people with depression21 and schizophrenia22, but recent RCTs of monoclonal antibodies targeting specific cytokine pathways have yielded null findings23–25. This highlights the need for strengthening causal inference using complementary techniques and data sources to inform appropriate selection of therapeutic target/agent in future trials. Furthermore, as existing studies have typically focused on a small number of immunological biomarkers, a comprehensive approach allowing investigations across hundreds of available biomarkers is necessary to obtain a more complete understanding of the role of immune dysfunction in neuropsychiatric conditions. We examined the causal influence of 735 genetically proxied immunological biomarkers on seven major neuropsychiatric conditions (schizophrenia, bipolar disorder, depression, anxiety, ADHD, autism, and Alzheimer’s disease), using cutting-edge genomic causal inference methods, MR and genetic colocalisation. We harnessed quantitative trait loci (QTL) data, capturing protein abundance (pQTL) and protein-coding gene expression (eQTL) in blood and brain, to gain mechanistic insights into potential systemic (blood) and brain-specific effects. We conducted a series of sensitivity analyses to examine the robustness of our findings, including the possibility of reverse causation. We used a systematic three-tier approach to appraise evidence of causality, by grouping biomarkers based on increasingly stringent criteria. Evidence of causality was complemented by the assessment of therapeutic tractability of identified causal biomarkers to inform future translation. ## Materials & Methods Please see Figure 1 for an overview. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F1) Figure 1. Analytic pipeline for the investigation of causal immunological biomarkers for neuropsychiatric conditions **Dorsolateral prefrontal cortex (DLPFC), **Cortex*. ### Genome-wide association studies (GWAS) of immunological biomarkers #### Blood plasma derived protein abundance (blood pQTLs) We used data from the largest genomic investigation of the human plasma proteome conducted in 34,557 European ancestry participants (discovery sample) of the UK Biobank (UKB)26 cohort, comprising 2,941 GWAS for 2,923 unique proteins assayed using the Olink Explore 3072 platform. Of these, we selected all proteins included in the Olink Inflammation panels I & II (n=735), which represent the most comprehensive collection of immunological biomarkers currently available (Supplementary Table 1). These 735 proteins formed the basis of our subsequent data extraction strategy from GWAS on blood and brain protein coding gene expression, and brain protein abundance. Details on the Olink Explore panels, assaying and genotyping in UKB can be found in the original publication26. #### Blood cell derived protein coding gene expression (blood eQTLs) We used data from the eQTLGEN Phase I27 study of blood-cell derived gene expression in a sample of 31,684 individuals. The study assessed the expression of 19,942 genes, including 561 immunological protein coding genes present in the Olink Inflammation panels I & II. #### Brain tissue derived protein abundance (brain pQTLs) To gain insights into potential brain specific effects, we additionally looked up the proteins of interest in BrainQTL, the most comprehensive investigation of the brain proteome currently available28. It includes levels of 7,376 proteins in the dorsolateral prefrontal cortex (DLPFC) of 330 individuals from the Religious Orders Study (ROS) and Memory and Ageing Projects (MAP). Brain pQTLs were available for 299 immunological proteins present in the Olink Inflammation panels I & II. #### Brain tissue derived protein coding gene expression (brain eQTLs) We used the latest meta-analysis of brain cortex gene expression (MetaBrain29) conducted in 6,601 RNA-seq samples (N=2,970) and covering 18,396 genes. This includes GWAS summary data for the expression of 715 genes encoding 715 proteins of interest in the brain cortex. Although MetaBrain provided data across different brain regions, the sample sizes were relatively small, and for this reason we used data for brain cortex rather than other brain regions (e.g., hippocampus, N= 168). Further details on the study samples, genotyping and QTL analyses from UKB, eQTLGEN, BrainQTL, and MetaBrain, can be found in the original publications26–29. ### GWAS of neuropsychiatric conditions We used the latest (at the time of analysis) available GWAS summary data on seven neuropsychiatric conditions. Specifically, two neurodevelopmental: autism30 (Ncases= 18,381, Ncontrols= 27,969), attention deficit hyperactivity disorder (ADHD)31 (Ncases= 38,691, Ncontrols= 186,843); four psychiatric: anxiety32 (Ncases= 7,016, Ncontrols= 14,745), depression33 (Ncases= 294,322, Ncontrols= 741,438), bipolar disorder34 (Ncases= 41,917, Ncontrols= 371,549), schizophrenia35 (Ncases= 76,755, Ncontrols= 243,649); and one neurodegenerative condition: Alzheimer’s disease36 (Ncases= 71,880, Ncontrols= 383,378). Details on the study samples, phenotype definition and genotyping can be found in the original publications30–36. ### Two-sample Mendelian randomization (MR) MR utilises the special properties of germline genetic variants to strengthen causal inference within observational data16. Here we implemented MR as an instrumental variables analysis using common genetic variants as instruments. The method can yield unbiased causal effect estimates under assumptions that the instruments should satisfy: (1) they must be associated with the exposure, (2) they must not be associated with any confounders of the exposure outcome associations, (3) they should operate on the outcome entirely through the exposure (i.e., no horizontal pleiotropy)37. For the present study, we performed two-sample MR, in which instrument-exposure and instrument-outcome effect sizes and standard errors were extracted from separate GWAS conducted in independent samples but representative of the same underlying population38. #### Instrument selection For each exposure (immunological protein abundance, protein-coding gene expression), we used as genetic instruments common genetic variants that met the genome-wide significance threshold (p≤5*10-08) and were independent (r2<0.01; 10,000 kb). We assessed the strength of each instrument by estimating their F-statistic (F≥10 indicates adequate instrument strength)39. Genetic instruments with an F-statistic <10 were excluded to minimise weak instrument bias. As instruments, we selected a total of 1,791 plasma pQTLs for 644 unique proteins; 1,474 blood cell-derived eQTLs for 501 protein coding genes; 32 DLPFC pQTLs for 32 proteins; 627 brain cortex eQTLs for 421 protein-coding genes. For blood pQTLs (UKB) and eQTLs (eQTLGEN), both *cis* and *trans* genetic instruments were available. Genetic instruments were categorised as *cis*-acting when they were located within proximity (±1Mb) to the gene regulatory region, and as *trans*-acting when located outside this window. Common genetic variants acting in *cis* to the protein-encoding gene are more likely to influence mRNA expression and protein levels (thus being less pleiotropic)40. On the other hand, *trans*-acting variants, are more likely to be pleiotropic due to their distance from the protein-encoding gene, but their inclusion can potentially increase the proportion of variance explained in the exposure, increasing the statistical power for MR analyses40,41. For brain pQTLs and eQTLs only *cis*-acting variants were used, because the BrainQTL study reported *cis*-pQTLs only. The MetaBrain study reported only the statistically significant *trans*-eQTLs, without information on the respective regions around them, rendering genetic colocalization analyses for the *trans*-acting variants impossible (details on genetic instruments used across analyses in Supplementary Tables 2-5). #### Statistical Analyses For each exposure, genetic instrument effect sizes and standard errors were extracted from each neuropsychiatric condition GWAS, and the variant-exposure, variant-outcome alleles were harmonised to ensure that effect sizes correspond to the same allele. If the exposure had only one associated variant, Wald ratio was used to generate causal effect estimates, and two-term Taylor expansion was used to approximate standard errors29,42. When more than one variant were available for an exposure, inverse variance weighted (IVW)43 regression was used. For IVW effect estimates, we also estimated Wald ratio estimates for each genetic instrument, to gain greater insight into the potential effects of individual variants (although these Wald ratio estimates were not prioritised in subsequent causal evidence appraisal, considering that the available multi-variant instruments for the respective IVW were likely to provide more reliable causal evidence). Details on the Wald ratio and IVW methods can be found in Supplementary Note 1. We used the Benjamini-Hochberg method44 to control false discovery rate (FDR) across our analyses (FDR<5%). In addition, we appraised findings using a strict Bonferroni-corrected p-value threshold 1.8*10-06 (0.05/27,468). ### Genetic colocalisation Colocalisation analysis can complement MR by elucidating a distinct aspect of the identified causal relationship between an exposure and an outcome45. Specifically, colocalisation allows the assessment of the hypothesis that any identified causal effects are driven by the same causal variant influencing both exposure and outcome, instead of distinct causal variants that are in linkage disequilibrium (LD) with each other46. In practice, the approach harnesses SNP coverage within the same specified locus for two traits of interest and tests whether the association signals for each trait at the locus are suggestive of a shared causal variant46. Considering that genetic instruments used in our analyses comprised either single variants (MR Wald ratio estimates) or multiple *cis* and/or *trans* variants (MR IVW estimates), we followed three distinct approaches for variant selection for colocalisation analyses to help prioritise (where possible) variants with the highest biological relevance for the exposure of interest. Specifically, when the instrument consisted of variants of which at least one was *cis*, the *cis* variant(s) was tested for colocalisation. If the instrument consisted of multiple *trans* variants, we used the *trans* variant with the smallest p-value for colocalisation. Finally, if the instrument was a single variant, the variant was tested for colocalisation regardless of whether it was *cis* or *trans*. We extracted regions within ±500KB around the instrumented variant and implemented the algorithm described by Robinson *et al*47 to perform pairwise conditional and colocalisation (PWCoCo) analysis, which assesses all conditionally independent signals in the exposure dataset region against all conditionally independent signals in the outcome data. Genotype data from mothers in the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort48 were used as the LD reference panel (N= 7,733; for ALSPAC cohort details and available genotype data see Supplementary Note 2). We ran these analyses using the default settings, as suggested by the authors in the original publications41,46. Evidence of colocalisation was considered if there was an H4 posterior probability of both traits having a shared causal variant ≥ 0.8, as proposed by the authors of the method. ### Steiger filtering We performed Steiger filtering to assess whether causal effect estimates were influenced by reverse causation49. The method assesses whether the genetic variants proxying the exposure explain more variance in the outcome, which, if true, suggests that the primary phenotype influenced by the variant is the outcome rather than the exposure. ### Drug target prioritisation and validation We used a three-tier system to prioritise evidence of causality for the biomarkers, and to explore their potential as drug targets. Tier A included findings that had evidence which passed the Bonferroni threshold (≤1.8*10-06), passed Steiger filtering, and passed colocalisation (H4≥0.8). Tier B included findings that passed the FDR threshold (<%5), passed Steiger filtering, and passed colocalisation (H4≥0.8). Additionally, for both Tiers A and B, the biomarker had to be proxied by a genetic instrument which was either a *cis* variant or in the case of multiple available variants, it included at least one *cis* variant. Tier C included findings where the MR estimates were from *trans* variants but fulfilled the same requirements as Tier B. When there was Tier A, B or C evidence for a biomarker across different QTLs (eQTL & pQTL) and tissue types (brain & blood), we performed genetic colocalisation analyses using PWCoCo between the QTLs of the biomarker. This approach allowed us to investigate whether the effects on the outcome were driven by the same underlying variant across QTLs and tissue types, which increases reliability of that molecular marker’s relationship with the condition50. We looked up potential therapeutic tractability of the identified Tier A, B or C biomarkers using small molecule, antibody binding, and/or any other treatment modality51 in the Open Targets Platform ([https://platform.opentargets.org/](https://platform.opentargets.org/)). The Open Targets Platform is a freely available online resource for drug target identification and prioritisation that integrates genetic and genomic data with existing evidence on protein structure and function, and information on approved drugs, and ongoing clinical trials51,52. Open Targets have categorised target tractability based on eight buckets/groups for small molecules and nine buckets/groups for antibodies (see [https://github.com/chembl/tractability\_pipeline_v2](https://github.com/chembl/tractability_pipeline_v2)). In order to aid interpretation, we categorised tractability into three mutually exclusive groups, in line with previous work53: Group 1. Strong druggability evidence: buckets 1, 2 & 3 for small molecules, antibodies, other modalities; Group 2. Likely or potentially druggable: buckets 4-8 for small molecules, 4 & 5 for antibodies; Group 3: Little or unknown druggability: remaining buckets. Data retrieved on 04/12/2023. ### Enriched pathways and phenotypes for the identified causal biomarkers To aid the interpretation of our findings and elucidate potential biological pathways underlying the identified causal biomarkers, we performed gene-set enrichment analyses using GeneNetwork54. Developed by the MetaBrain consortium, GeneNetwork allows enrichment analyses using terms available on the Gene Ontology (GO), KEGG and Reactome pathway resources, as well as the Human Phenotype Ontology (HPO) database. We used the GeneNetwork browser (analyses conducted on 12/12/2023) and entered in the analyses the biomarkers that satisfied the Tier A, B and C criteria for each neuropsychiatric condition. These analyses were conducted only for neuropsychiatric conditions with at least five identified causal biomarkers (satisfying Tier A, B, or C criteria). ### Bi-directional two-sample MR To assess reverse causation, we tested the causal effects of genetic liability to each neuropsychiatric condition on circulating immunological proteins. Genetic instruments for each condition were extracted from the respective GWAS using a p-value threshold of ≤5*10-08 (r2<0.01; 10,000 kb). The only exceptions were autism and anxiety, for which a p-value threshold of ≤5*10-07 was used as there were insufficient instruments at the genome-wide significance threshold (2 and 1 instrument respectively). Details on the instruments for each neuropsychiatric condition can be found in Supplementary Table 7. Genetic instruments were then extracted from the GWAS of the immunological proteins (735 proteins)26 and their alleles were harmonised. Causal effects were estimated using the IVW approach (Supplementary Note 1)43. Due to the number of tests conducted (7 phenotypes*735 immunological biomarkers available in UKB) we used Bonferroni correction (p≤9.7*10-06). ### Software Analyses were carried out using the computational facilities of the Advanced Computing Research Centre of the University of Bristol ([http://www.bris.ac.uk/acrc/](http://www.bris.ac.uk/acrc/)). Blood plasma pQTL data and blood cell derived eQTL data were extracted and processed using the gwasvcf package version 1.0 in R ([https://github.com/MRCIEU/gwasvcf](https://github.com/MRCIEU/gwasvcf))55. Two-sample MR, Steiger filtering, and bi-directional MR analyses were conducted using functions from the TwoSampleMR R package version 0.5.6 ([https://github.com/MRCIEU/TwoSampleMR](https://github.com/MRCIEU/TwoSampleMR))56 and the mrpipeline R package ([https://github.com/jwr-git/mrpipeline](https://github.com/jwr-git/mrpipeline)). The PWCoCo algorithm was implemented using the Pair-Wise Conditional analysis and Colocalisation analysis package v1.0 ([https://github.com/jwr-git/pwcoco](https://github.com/jwr-git/pwcoco))47. Pathway and phenotype enrichment analyses were conducted using the GeneNetwork browser v2.0 ([https://www.genenetwork.nl/](https://www.genenetwork.nl/))54. ### Data availability Across all analyses published summary-level data were used and no patient identifiable information was included. UKB blood pQTL data can be accessed through the portal: [http://ukb-ppp.gwas.eu](http://ukb-ppp.gwas.eu). deCODE blood pQTL data can be accessed through the platform: [https://www.decode.com/summarydata/](https://www.decode.com/summarydata/). Brain pQTL data can be accessed through the Synapse portal: [https://www.synapse.org/#!Synapse:syn24172458](https://www.synapse.org/#!Synapse:syn24172458). Blood eQTL data can be accessed through [https://www.eqtlgen.org/phase1.html](https://www.eqtlgen.org/phase1.html). Brain cortex eQTL data can be accessed through the MetaBrain platform: [https://www.metabrain.nl/](https://www.metabrain.nl/). GWAS data on ADHD, autism, anxiety, bipolar disorder, and schizophrenia can be accessed at: [https://pgc.unc.edu/for-researchers/download-results/](https://pgc.unc.edu/for-researchers/download-results/). GWAS data on depression can be accessed at: [https://ipsych.dk/en/research/downloads/](https://ipsych.dk/en/research/downloads/). GWAS data on Alzheimer’s disease can be accessed at: [https://ctg.cncr.nl/software/summary_statistics](https://ctg.cncr.nl/software/summary_statistics). ### Code availability A version of the code at time of publication will be available at the study-dedicated GitHub repository. ## Results ### Immunological drivers for neuropsychiatric conditions In total, we found evidence for 270 potentially causal relationships corresponding to 122 unique immunological biomarkers that passed the FDR<5% (Supplementary Table 7). Among these 21 unique biomarkers met our strict Tier A, B or C criteria for causal evidence. One biomarker met the most stringent Tier A criteria (i.e., passed the stringent Bonferroni threshold:1.8*10-06, Steiger filtering, was supported by evidence of colocalisation, and the instrument consisted of *cis* variants). Twenty-one biomarkers (4 unique proteins and 17 genes) met Tier B criteria (i.e., passed the FDR threshold, Steiger filtering, were supported by evidence of colocalisation, and the instruments consisted of *cis* variants). Six effects corresponding to 4 unique proteins met Tier C criteria (genetic instrument consisted of *trans* variants and fulfilled the Tier B requirements). Schizophrenia (n=64), depression (n=33), bipolar disorder (n=31) and Alzheimer’s disease (n=23) had the highest number of potentially causal immunological markers after FDR correction. ### Condition specific findings #### Neurodevelopmental conditions For autism, we identified 14 potentially causal immunological biomarkers after FDR correction, two of which met Tier B/C criteria. The effect of genetically proxied expression of *ANXA1* in blood tissue (FDR= 0.002; H4>0.8) fulfilled the Tier B criteria. Genetically proxied levels of CEBPA in blood fulfilled the Tier C criteria (FDR=0.01, H4>0.8). For ADHD, 9 potentially causal immunological biomarkers were identified after FDR, of which genetically proxied expression of *GCHFR* in blood fulfilled the Tier B criteria (FDR=0.006; H4>0.8). The results are visualised in Figures 2, 3, and detailed MR and colocalisation findings are available in Supplementary Tables 7 and 8. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F2) Figure 2. Volcano plot illustrating the combined results of the analyses investigating the causal effects of genetically proxied immunological biomarkers on autism *Red dashed lines denote the FDR threshold, and red dotted lines denote the Bonferroni threshold*. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F3) Figure 3. Volcano plot illustrating the combined results of the analyses investigating the causal effects of genetically proxied immunological biomarkers on ADHD *Red dashed lines denote the FDR threshold, and red dotted lines denote the Bonferroni threshold*. #### Psychotic and affective disorders We found 64 potentially causal immunological biomarkers for schizophrenia after FDR correction, of which 8 met the Tier B criteria. Specifically, we found evidence of causal effects for genetically proxied expression of *ACE* in blood (FDR=0.008; H4>0.8) and brain cortex (FDR=0.002; H4>0.8), genetically proxied expression of *PDIA3* (FDR= 3*10-05; H4>0.8) in blood, genetically proxied expression of *TNFRSF17* (FDR=0.02; H4>0.8), *CD40* (FDR=0.004; H4>0.8), *SERPINI1* (FDR=0.05; H4>0.8), *EVI5* (FDR=0.003, H4>0.8) in brain cortex, and genetically proxied levels of RABEP1 (FDR= 0.01; H4>0.8), and DNER (FDR=0.008; H4>0.8) in blood. Furthermore, a blood *trans* pQTL of BTN2A1 (FDR= 2*10-15; H4>0.8) and a blood *trans* pQTL of MYOM3 (FDR=0.01, H4>0.8) fulfilled the Tier C criteria. See Figure 4, Supplementary Tables 7 and 8. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F4) Figure 4. Volcano plot illustrating the combined results of the analyses investigating the causal effects of genetically proxied immunological biomarkers on schizophrenia *Red dashed lines denote the FDR threshold, and red dotted lines denote the Bonferroni threshold*. Among the 33 potentially causal immunological biomarkers identified for depression, genetically proxied expression of *AMN* (FDR=0.03, H4<0.8) and *EP300* (FDR=2*10 -06) in blood and genetically proxied expression of *FCN1* (FDR=0.02, H4>0.8) and *LATS1* (FDR=0.006; H4>0.8) in the brain cortex fulfilled the Tier B criteria. In addition, genetically proxied levels of BTN2A1 in blood (FDR=4*10-06; H4>0.8) fulfilled the Tier C criteria. See Figure 5, Supplementary Tables 7 and 8. ![Figure 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F5.medium.gif) [Figure 5.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F5) Figure 5. Volcano plot illustrating the combined results of the analyses investigating the causal effects of genetically proxied immunological biomarkers on depression *Red dashed lines denote the FDR threshold, and red dotted lines denote the Bonferroni threshold*. For bipolar disorder, we found 31 potentially causal biomarkers. Among these, genetically proxied levels of CD40 (FDR=0.01; H4>0.8) in blood and genetically proxied expression of *CD40* in the brain cortex (FDR=0.002, H4>0.8), as well as genetically proxied expression of *DNPH1* (FDR=0.04; H4>0.8) in blood fulfilled the Tier B criteria. Genetically proxied levels of BTN2A1 in blood (FDR=3*10-05, H4>0.8) fulfilled the Tier C criteria (Figure 6, Supplementary Tables 7 and 8). ![Figure 6.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F6.medium.gif) [Figure 6.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F6) Figure 6. Volcano plot illustrating the combined results of the analyses investigating the causal effects of genetically immunological biomarkers on bipolar disorder *Red dashed lines denote the FDR threshold, and red dotted lines denote the Bonferroni threshold*. No estimated causal effects of genetically proxied immunological biomarkers on anxiety surpassed the FDR threshold (<5%; Supplementary Tables 7 and 8). #### Alzheimer’s disease We identified 23 potentially causal biomarkers for Alzheimer’s disease. Among these, genetically proxied expression of *CR1* (FDR= 4*10-14; H4>0.8) in the brain cortex fulfilled the Tier A criteria. In addition, genetically proxied expression of *ACE* in blood (FDR= 0.006; H4>0.8) and brain (FDR=0.001; H4>0.8) as well as genetically proxied levels of APOE in blood (FDR= 3*10-06; H4>0.8) fulfilled the Tier B criteria. Genetically proxied levels of KLRB1 (FDR=0.001, H4>0.8) fulfilled the Tier C criteria (Figure 7, Supplementary Tables 7 and 8). ![Figure 7.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F7.medium.gif) [Figure 7.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F7) Figure 7. Volcano plot illustrating the combined results of the analyses investigating the causal links between genetically proxied immunological biomarkers on Alzheimer’s disease *Red dashed lines denote the FDR threshold, and red dashed lines denote the Bonferroni threshold*. ### Drug target identification, prioritisation, and validation In total, 18 unique biomarkers meeting our strict Tier A, B or C criteria for causal evidence are therapeutically tractable (Table 1). Notably, *ACE*, which had Tier B evidence for both schizophrenia and Alzheimer’s, has approved drugs for cardiovascular indications. *CD40* (Tier B evidence for both schizophrenia and bipolar disorder) has drugs in clinical trials. Furthermore, *TNFRSF17* (Tier B evidence for schizophrenia) has approved drugs. See Supplementary Table 9 for details on drugs approved or in clinical trials for *ACE*, *CD40* and *TNFRSF17*. View this table: [Table 1.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/T1) Table 1. Therapeutic tractability of immunological genes identified as causal for neuropsychiatric conditions. Importantly, the effects of *ACE* on schizophrenia and Alzheimer’s were supported by both blood and brain eQTLs. The effects of *CD40* on bipolar disorder were supported by a blood pQTL and a brain eQTL. The brain eQTL was also causal for schizophrenia (Figure 8a). We performed colocalisation analyses between the blood and brain eQTLs for *ACE* as well as the blood pQTLs and brain eQTL for *CD40*. We found evidence suggesting that the effects of *ACE* were driven by a shared underlying variant (H4=0.91; Figure 8b), whereas the effects of *CD40* were driven by distinct causal variants (H3=0.99). ![Figure 8a.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F8.medium.gif) [Figure 8a.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F8) Figure 8a. Visual summary of the MR evidence suggesting causal effects of CD40 blood pQTLs and brain eQTLs on bipolar disorder and schizophrenia, as well as ACE brain and blood eQTLs on Alzheimer’s disease and schizophrenia. ![Figure 8b.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F9.medium.gif) [Figure 8b.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F9) Figure 8b. Locus plot of the ACE brain and blood eQTLs. ### Enriched pathways and phenotypes for the identified causal biomarkers Analyses were conducted for schizophrenia and depression, as these had enough (≥5) causal biomarkers satisfying the Tier A, B, or C criteria. For schizophrenia, 10 biomarkers were entered in GeneNetwork. The strongest evidence of enrichment (p≤2*10-05) was for reactome pathways belonging to the “metabolism of RNA” superpathway, which has been implicated in a number of neurological conditions57. We also found strong enrichment for HPO developmental phenotypes related to fetal abnormalities including congenital malformation of the great arteries (p=3*10-04) and thickened nuchal skin fold (p=2*10-03). For depression, the strongest evidence of enrichment (p≤6*10-04) was for reactome pathways belonging to the “DNA repair” superpathway. This superpathway has been implicated in cancer58. Similarly, there was strong evidence of enrichment for HPO cancer phenotypes. See Supplementary Table 10 for all enrichment findings, and Figure 9 for the five most significantly enriched pathways and phenotypes for schizophrenia and depression related biomarkers. ![Figure 9.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/02/17/2024.02.16.24302885/F10.medium.gif) [Figure 9.](http://medrxiv.org/content/early/2024/02/17/2024.02.16.24302885/F10) Figure 9. Top five enriched pathways and phenotypes for causal immunological biomarkers identified in schizophrenia and depression ### Evidence of reverse causation In bidirectional MR analyses, none of the estimated causal effects of genetic liability to autism, ADHD, schizophrenia, bipolar disorder, or anxiety on levels of immunological proteins, surpassed the Bonferroni correction threshold (p≤9.7*10-06). However, we found evidence of causal effects of genetic liability to depression on levels of CXCL17 (p=5*10-08), PRSS8 (p=7*10-07) and TNFRSF13B (p=8*10-06). Similarly, we found that genetic liability to Alzheimer’s disease had causal effects on levels of APOA2 (p=8*10-09), APOE (p=1*10-18), APOF (p=6*10-12), ENPP7 (p=5*10-07), IL32 (p=3*10-11), C1orf56 (p=5*10-15), SMPDL3A (p=1*10-07), VNN1 (p=5*10-06). See Supplementary Table 11. ## Discussion Recent decades have seen limited progress in new therapeutics for neuropsychiatric conditions. Despite converging evidence implicating immune dysfunction in several neuropsychiatric conditions, the success of immunotherapy clinical trials remains elusive. One key barrier is the lack of a clear understanding of causality to inform appropriate selection of therapeutic target/agent. In this study, using cutting-edge genomic causal inference methods applied to largescale proteomic and gene expression data from blood and brain, we have assessed evidence for causality for the largest available selection of immune-response related biomarkers in relation to seven neuropsychiatric conditions. We provide robust evidence for causality for 21 immunological biomarkers confirming that both brain specific and systemic immune response contribute to pathogenesis of neuropsychiatric conditions, especially schizophrenia, Alzheimer’s disease, depression, and bipolar disorder. Our work highlights the potential opportunity for repurposing existing drugs for neuropsychiatric indications. ### Novel immunological biomarkers Among putative causal biomarkers identified, two related to depression are novel. Specifically, *LATS1* (6q25.1) and *FCN1* (9q34.3) are part of a broad network of genes related to type I interferon (IFN-I) signalling59,60, which is implicated in inhibition of viral infections and tumour growth suppression61. Our finding is consistent with induction of depression in hepatitis C patients following treatment with interferon alpha6, which binds to and activates IFN-1 receptors. The potential IFN-1 mediated tumour related effect of these genes are in line with our gene-set enrichment findings indicating significant enrichment for cancer-related phenotypes. Of note, our results correspond to *LATS1* and *FCN1* expression in brain cortex, not blood. IFN-I type signalling in brain has been implicated in brain aging, Alzheimer’s disease, and multiple sclerosis62,63. Therefore, the possibility of distinct tissue/organ specific effects of these genes for different conditions requires further investigation. ### From causal biomarkers to drug targets for neuropsychiatric conditions Identifying causal immunological biomarkers for neuropsychiatric conditions is a pivotal step toward developing new or repurposing existing interventions. We found that 18 of the identified biomarkers are potentially druggable. Three of the identified biomarkers, *ACE* (effects on schizophrenia & Alzheimer’s), *CD40* (effects on schizophrenia & bipolar), *TNFRSF17* (effects on schizophrenia) have drugs approved or in advanced clinical trials for several indications including cardiovascular and autoimmune conditions, which provides an opportunity for drug repurposing. However, there are important considerations regarding therapeutics development which we discuss below using our findings for schizophrenia (for which we identified the highest number of causal biomarkers) as an example. #### Pathways from transcription to translation The effects on schizophrenia for more than half of the 10 immunological biomarkers with Tier B or C evidence for causality identified were through genetic variants linked to gene expression (*TNFRSF17*, *CD40*, *SERPINI1*, *EVI5, ACE, PDIA3*) but not respective protein abundance. This could be a result of limited statistical power, as the brain pQTL dataset had a total sample size of 330 individuals. However, another potential explanation may be alternative splicing events. Alternative splicing has a central role in the pathway from transcription to translation as it results in the production of multiple proteins via different signalling pathways64. Alternative splicing events may play an important role in schizophrenia aetiopathogenesis65. Therefore, further studies with larger sample sizes for brain pQTL and studies using genomic data on alternative splicing (sQTLs) are required. #### Tissue specific effects Half of the identified biomarkers had effects on schizophrenia via gene expression in the brain cortex (*TNFRSF17*, *CD40*, *SERPINI1*, *EVI5, ACE*), suggesting that both systemic and brain-specific immunological processes are important for its aetiopathogenesis66. Particularly for *ACE,* the identified MR effects were supported by both blood and brain cortex eQTLs and the estimates were directionally consistent across both tissues. Furthermore, colocalisation analyses suggested that the effects were likely to be driven by the same underlying variant, increasing the confidence regarding a causal role of ACE in schizophrenia. Now interventional studies are required to assess the therapeutic potential of targeting ACE for schizophrenia. Epidemiological studies suggest associations of immune-mediated respiratory and gastrointestinal conditions with schizophrenia (e.g., asthma67, inflammatory bowel disease68). As *ACE* is expressed primarily in lung and small intestine69, our findings also highlight the potential relevance of the lung-brain or gut-brain axis for schizophrenia pathogenesis70. #### Effects across neuropsychiatric conditions We found that decreased expression of *ACE* in blood and brain cortex is linked to increased risk of both schizophrenia and Alzheimer’s disease. This is consistent with results from previous MR studies71,72. Considering that ACE inhibitors are widely used for the management of hypertension, these findings require further investigation. The identified effect for Alzheimer’s particularly may be a result of survival bias, considering that hypertension can lead to early mortality and therefore individuals may not live long enough to be diagnosed with the condition73,74. Beyond its effects on hypertension, ACE inhibition in rats leads to memory and learning impairments75. Therefore, another possibility is that ACE inhibition does not causally influence risk to the conditions per se, but some of their common phenotypic expressions, such as cognitive decline, which is common to both schizophrenia and Alzheimer’s disease. Therefore, choosing the right outcome would be as important as choosing the right drug target in future RCTs. Similarly, *CD40* expression in brain may influence risk of both schizophrenia and bipolar disorder by causally influencing psychotic symptoms, which are common to both conditions. These possibilities require further investigation. #### Effects on developmental stage and progression Our study design allowed us to investigate the causal effects of immunological biomarkers on the onset of neuropsychiatric conditions but not progression76. Identifying actionable treatment targets can be particularly complex for conditions with neurodevelopmental origins such as schizophrenia, where pathogenic changes could take place well before the emergence of clinical symptoms77. Our gene-set enrichment analyses show significant enrichment for early developmental phenotypes related to fetal abnormalities, suggesting that immune-mediated neurodevelopmental alterations could contribute to schizophrenia. Therefore, the question of the potential utility of the identified targets in conditions with neurodevelopmental origins remains, and requires careful consideration of conceptual, methodological, and ethical aspects. ### Limitations Our study has some methodological limitations. First, the study was conducted using GWAS data of European ancestry individuals, and therefore generalisability of our findings to other populations remains a concern. Second, although we used the largest GWAS data available the possibility of limited statistical power cannot be excluded, particularly for brain pQTL data, and the anxiety GWAS. Third, our enrichment analyses were based on a small number of causal biomarkers (10 in schizophrenia and 5 in depression), which may limit the reliability of these findings. Fourth, UKB proteomic GWAS had some sample overlap with some of the neuropsychiatric conditions, notably Alzheimer’s disease (>50%), depression (34%), and bipolar disorder (14%). Sample overlap can introduce a bias toward the observational estimate78, though this is unlikely to adversely influence hypothesis testing79 and we observed the highest number of associations for schizophrenia which had limited overlap with the proteomic GWAS sample. Fifth, though our results are strongly suggestive of causal relationship we are unable to prove causality due to potential horizontal pleiotropy or violations of other MR assumptions such as gene-environment equivalence and consistency of treatment effects. Interventional studies are required for the ultimate proof of causal relationship, which our findings should inform. ## Conclusions Using a comprehensive analytic approach allowing the integration of genomic data on protein and gene expression across blood and brain, we identify causal effects for 21 immunological biomarkers on seven neuropsychiatric conditions. Of these, 18 are therapeutically tractable suggesting that they could be potential targets for neuropsychiatric indications. Further validation of the identified biomarkers through interventional studies and understanding the mechanisms underlying these effects are now necessary for greater insight into pathogenesis of neuropsychiatric conditions and for developing new treatments to improve the lives of people with these conditions. ## Conflicts of interest JWR is a full-time employee of Boehringer Ingelheim but undertook all relevant work when at the University of Bristol. TGR is a full-time employee of GlaxoSmithKline outside of this work. No funding body has influenced data collection, analyses, or their interpretation. ## Funding Statement This work is funded by a UK Medical Research Council (MRC) grant to GMK via the Integrative Epidemiology Unit (IEU) at the University of Bristol (MC\_UU\_00032/06). This grant also supports CD. GMK acknowledges additional funding support from the Wellcome Trust (201486/Z/16/Z and 201486/B/16/Z), MRC (MR/W014416/1; MR/S037675/1) and the UK National Institute of Health and Care Research (NIHR) Bristol Biomedical Research Centre (NIHR 203315). GMK, TRG and AMM acknowledge funding support from the MRC (The CHECKPOINT Hub, APP4735-GTEE-2024). AMM is supported by the Wellcome Trust (220857/Z/20/Z) and UKRI (MR/W014386/1). GDS, HJ, DR, GH, TRG, and GMK are supported by the National Institute for Health and Care Research Bristol Biomedical Research Centre. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care. RG acknowledges funding support from the Swedish Research Council (VR2017-02900; 2022-00592). AH was supported by grants from the South-Eastern Norway Regional Health Authority (2020022, 2018059) and the Research Council of Norway (274611, 288083, 336085). The Medical Research Council (MRC) and the University of Bristol support the MRC Integrative Epidemiology Unit (MC\_UU\_00011/1, MC\_UU_00011/3, MC_UU_00011/5). This research was funded in part, by the Wellcome Trust. For the purpose of Open Access, the author has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising from this submission. The UK Medical Research Council and Wellcome (Grant ref: 217065/Z/19/Z) and the University of Bristol provide core support for ALSPAC. This publication is the work of the authors and CD, GMK will serve as guarantors for the contents of this paper. Genomewide genotyping data was generated by Sample Logistics and Genotyping Facilities at Wellcome Sanger Institute and LabCorp (Laboratory Corporation of America) using support from 23andMe. ## Ethics Statement ALSPAC data used as reference panel: ethical approval was obtained from the ALSPAC Ethics and Law Committee and the Local Research Ethics Committees. We are extremely grateful to all the families who took part in this study, the midwives for their help in recruiting them, and the whole ALSPAC team, which includes interviewers, computer and laboratory technicians, clerical workers, research scientists, volunteers, managers, receptionists, and nurses. GWAS data: across all analyses summary data were used. These data are publicly available for research purposes. Details on the ethics declarations for each GWAS study can be found in the original publications. ## Supporting information Supplementary Material [[supplements/302885_file03.xlsx]](pending:yes) ## Data Availability All GWAS data used in this work is publicly available for research purposes. Individual-level data from the ALSPAC birth cohort are not publicly available for reasons of clinical confidentiality. Data can be accessed after application to the ALSPAC Executive Team who will respond within 10 working days. Application instructions and data use agreements are available at [http://www.bristol.ac.uk/alspac/researchers/access/](http://www.bristol.ac.uk/alspac/researchers/access/). * Received February 16, 2024. * Revision received February 16, 2024. * Accepted February 17, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## References 1. 1.GBD 2019 Mental Disorders Collaborators. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. The Lancet Psychiatry (2022). 2. 2.Vos, T. et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet 390, 1211–1259 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=doi:10.1016/S0140-6736(17)32154-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28919117&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 3. 3.Bray, N. J. & O’Donovan, M. C. The genetics of neuropsychiatric disorders. Brain Neurosci. Adv. 2, 2398212818799271 (2018). 4. 4.Howes, O. D., Thase, M. E. & Pillinger, T. Treatment resistance in psychiatry: state of the art and new directions. Mol. Psychiatry 1–15 (2021). 5. 5.Kupferschmidt, K. In new strategy, Wellcome Trust takes on global health concerns. (2020). 6. 6.Udina, M. et al. Interferon-induced depression in chronic hepatitis C: a systematic review and meta-analysis. J. Clin. Psychiatry 73, 1128–1138 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4088/JCP.12r07694&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22967776&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 7. 7.Harrison, N. A. et al. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol. Psychiatry 66, 407– 414 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2009.03.015&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19423079&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000269330900002&link_type=ISI) 8. 8.Goldsmith, D. R., Rapaport, M. H. & Miller, B. J. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol. Psychiatry 21, 1696 (2016). 9. 9.Wang, A. K. & Miller, B. J. Meta-analysis of Cerebrospinal Fluid Cytokine and Tryptophan Catabolite Alterations in Psychiatric Patients: Comparisons Between Schizophrenia, Bipolar Disorder, and Depression. Schizophr. Bull. 44, 75–83 (2018). 10. 10.Setiawan, E. et al. Role of Translocator Protein Density, a Marker of Neuroinflammation, in the Brain During Major Depressive Episodes. JAMA Psychiatry 72, 268–275 (2015). 11. 11.Benros, M. E. et al. A nationwide study on the risk of autoimmune diseases in individuals with a personal or a family history of schizophrenia and related psychosis. Am. J. Psychiatry 171, 218–226 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2013.13010086&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24129899&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000330601100016&link_type=ISI) 12. 12.Dalman, C. et al. Infections in the CNS during childhood and the risk of subsequent psychotic illness: a cohort study of more than one million Swedish subjects. Am. J. Psychiatry 165, 59–65 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2007.07050740&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18056223&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000252186000014&link_type=ISI) 13. 13.Hegvik, T.-A. et al. Familial co-aggregation of attention-deficit/hyperactivity disorder and autoimmune diseases: a cohort study based on Swedish population-wide registers. Int. J. Epidemiol. 51, 898–909 (2022). 14. 14.Sun, J. et al. Hospital-treated infections in early- and mid-life and risk of Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis: A nationwide nested case-control study in Sweden. PLOS Med. 19, e1004092 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1004092&link_type=DOI) 15. 15.Benros, M. E. & Mortensen, P. B. Role of infection, autoimmunity, atopic disorders, and the immune system in schizophrenia: evidence from epidemiological and genetic studies. Neuroinflammation Schizophr. 141–159 (2019). 16. 16.Sanderson, E. et al. Mendelian randomization. Nat. Rev. Methods Prim. 2, 1–21 (2022). 17. 17.Davey Smith, G. & Hemani, G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum. Mol. Genet. 23, R89–R98 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/hmg/ddu328&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25064373&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000349825700013&link_type=ISI) 18. 18.Holmes, M. V, Richardson, T. G., Ference, B. A., Davies, N. M. & Davey Smith, G. Integrating genomics with biomarkers and therapeutic targets to invigorate cardiovascular drug development. Nat. Rev. Cardiol. 18, 435–453 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41569-020-00493-1&link_type=DOI) 19. 19.Hartwig, F. P., Borges, M. C., Horta, B. L., Bowden, J. & Davey Smith, G. Inflammatory biomarkers and risk of schizophrenia: a 2-sample mendelian randomization study. JAMA psychiatry 74, 1226–1233 (2017). 20. 20.Perry, B. I. et al. Associations of immunological proteins/traits with schizophrenia, major depression and bipolar disorder: A bi-directional two-sample mendelian randomization study. Brain. Behav. Immun. 97, 176–185 (2021). 21. 21.Köhler-Forsberg, O. et al. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: meta-analysis of clinical trials. Acta Psychiatr. Scand. 139, 404–419 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/acps.13016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30834514&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 22. 22.Cho, M. et al. Adjunctive use of anti-inflammatory drugs for schizophrenia: A meta-analytic investigation of randomized controlled trials. Aust. N. Z. J. Psychiatry 53, 742–759 (2019). 23. 23.McIntyre, R. S. et al. Efficacy of Adjunctive Infliximab vs Placebo in the Treatment of Adults With Bipolar I/II Depression: A Randomized Clinical Trial. JAMA psychiatry 76, 783–790 (2019). 24. 24.Raison, C. L. et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA psychiatry 70, 31–41 (2013). 25. 25.Girgis, R. R. et al. A Randomized, Double-Blind, Placebo-Controlled Clinical Trial of Tocilizumab, An Interleukin-6 Receptor Antibody, For Residual Symptoms in Schizophrenia. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 43, 1317–1323 (2018). 26. 26.Sun, B. B. et al. Plasma proteomic associations with genetics and health in the UK Biobank. Nature 622, 329–338 (2023). 27. 27.Võsa, U. et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 53, 1300– 1310 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0022146515594631.Marriage&link_type=DOI) 28. 28.Robins, C. et al. Genetic control of the human brain proteome. Am. J. Hum. Genet. 108, 400–410 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2021.01.012&link_type=DOI) 29. 29.de Klein, N. et al. Brain expression quantitative trait locus and network analyses reveal downstream effects and putative drivers for brain-related diseases. Nat. Genet. 1–12 (2023). 30. 30.Grove, J. et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 1 (2019). 31. 31.Demontis, D. et al. Genome-wide analyses of ADHD identify 27 risk loci, refine the genetic architecture and implicate several cognitive domains. Nat. Genet. 55, 198– 208 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/S41588-022-01285-8&link_type=DOI) 32. 32.Otowa, T. et al. Meta-analysis of genome-wide association studies of anxiety disorders. Mol. Psychiatry 21, 1391–1399 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/mp.2015.197&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26754954&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 33. 33.Als, T. D. et al. Depression pathophysiology, risk prediction of recurrence and comorbid psychiatric disorders using genome-wide analyses. Nat. Med. 29, 1832– 1844 (2023). 34. 34.Mullins, N. et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat. Genet. 53, 817–829 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-021-00857-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34002096&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 35. 35.Trubetskoy, V. et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 604, 502–508 (2022). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 36. 36.Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 51, 404–413 (2019). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 37. 37.Haycock, P. C. et al. Best (but oft-forgotten) practices: the design, analysis, and interpretation of Mendelian randomization studies. Am. J. Clin. Nutr. 103, 965–978 (2016). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiYWpjbiI7czo1OiJyZXNpZCI7czo5OiIxMDMvNC85NjUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMi8xNy8yMDI0LjAyLjE2LjI0MzAyODg1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 38. 38.Burgess, S. et al. Using published data in Mendelian randomization: a blueprint for efficient identification of causal risk factors. Eur. J. Epidemiol. 30, 543–552 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10654-015-0011-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25773750&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 39. 39.Burgess, S., Thompson, S. G. & collaboration, C. R. P. C. H. D. genetics. Avoiding bias from weak instruments in Mendelian randomization studies. Int. J. Epidemiol. 40, 755–764 (2011). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyr036&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21414999&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000293618300027&link_type=ISI) 40. 40.Swerdlow, D. I. et al. Selecting instruments for Mendelian randomization in the wake of genome-wide association studies. Int. J. Epidemiol. 45, 1600–1616 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyw088&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27342221&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 41. 41.Zheng, J. et al. Phenome-wide Mendelian randomization mapping the influence of the plasma proteome on complex diseases. Nat. Genet. 52, 1122–1131 (2020). 42. 42.Wald, A. The fitting of straight lines if both variables are subject to error. Ann. Math. Stat. 11, 284–300 (1940). 43. 43.Bowden, J., Davey Smith, G. & Burgess, S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int. J. Epidemiol. 44, 512–525 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyv080&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26050253&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 44. 44.Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B 57, 289–300 (1995). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0748730414553029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25326247&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 45. 45.Burgess, S., Foley, C. N. & Zuber, V. Inferring causal relationships between risk factors and outcomes from genome-wide association study data. Annu. Rev. Genomics Hum. Genet. 19, 303–327 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1146/annurev-genom-083117-021731&link_type=DOI) 46. 46.Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pgen.1004383&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24830394&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 47. 47.Robinson, J. W., et al. An efficient and robust tool for colocalisation: Pair-wise Conditional and Colocalisation (PWCoCo). bioRxiv 2022.08.08.503158 (2022) doi:10.1101/2022.08.08.503158. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMi4wOC4wOC41MDMxNTh2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAyLzE3LzIwMjQuMDIuMTYuMjQzMDI4ODUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 48. 48.Fraser, A. et al. Cohort profile: the Avon Longitudinal Study of Parents and Children: ALSPAC mothers cohort. Int. J. Epidemiol. 42, 97–110 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dys066&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22507742&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316699300011&link_type=ISI) 49. 49.Hemani, G., Tilling, K. & Davey Smith, G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 13, e1007081 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pgen.1007081&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29149188&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 50. 50.Robinson, J. W., et al. Evaluating the potential benefits and pitfalls of combining protein and expression quantitative trait loci in evidencing drug targets. bioRxiv (2022). 51. 51.Brown, K. K. et al. Approaches to target tractability assessment - a practical perspective. Medchemcomm 9, 606–613 (2018). 52. 52.Koscielny, G. et al. Open Targets: a platform for therapeutic target identification and validation. Nucleic Acids Res. 45, D985–D994 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkw1055&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27899665&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 53. 53.Rasooly, D. et al. Genome-wide association analysis and Mendelian randomization proteomics identify drug targets for heart failure. medRxiv 2022.04.14.22273877 (2023) doi:10.1101/2022.04.14.22273877. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMi4wNC4xNC4yMjI3Mzg3N3YzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMTcvMjAyNC4wMi4xNi4yNDMwMjg4NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 54. 54.Pers, T. H. et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 6, 5890 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ncomms6890&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25597830&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 55. 55.Lyon, M. S. et al. The variant call format provides efficient and robust storage of GWAS summary statistics. Genome Biol. 22, 32 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-020-02248-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33441155&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 56. 56.Walker, V. M. et al. Using the MR-Base platform to investigate risk factors and drug targets for thousands of phenotypes. Wellcome open Res. 4, (2019). 57. 57.Nussbacher, J. K., Tabet, R., Yeo, G. W. & Lagier-Tourenne, C. Disruption of RNA metabolism in neurological diseases and emerging therapeutic interventions. Neuron 102, 294–320 (2019). 58. 58.Helleday, T., Petermann, E., Lundin, C., Hodgson, B. & Sharma, R. A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 8, 193–204 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrc2342&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18256616&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253407800014&link_type=ISI) 59. 59.Fineschi, S. et al. Inflammation and Interferon Signatures in Peripheral B-Lymphocytes and Sera of Individuals With Fibromyalgia. Front. Immunol. 13, 874490 (2022). 60. 60.Zuo, Y. et al. LATS1 is a central signal transmitter for achieving full type-I interferon activity. Sci. Adv. 8, eabj3887 (2022). 61. 61.Yu, R., Zhu, B. & Chen, D. Type I interferon-mediated tumor immunity and its role in immunotherapy. Cell. Mol. Life Sci. 79, 191 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00018-022-04219-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35292881&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 62. 62.Paul, S., Ricour, C., Sommereyns, C., Sorgeloos, F. & Michiels, T. Type I interferon response in the central nervous system. Biochimie 89, 770–778 (2007). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biochi.2007.02.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17408841&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248502100010&link_type=ISI) 63. 63.Deczkowska, A., Baruch, K. & Schwartz, M. Type I/II interferon balance in the regulation of brain physiology and pathology. Trends Immunol. 37, 181–192 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.it.2016.01.006&link_type=DOI) 64. 64.Kornblihtt, A. R. et al. Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat. Rev. Mol. Cell Biol. 14, 153–165 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrm3525&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23385723&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 65. 65.Zhang, C.-Y., Xiao, X., Zhang, Z., Hu, Z. & Li, M. An alternative splicing hypothesis for neuropathology of schizophrenia: evidence from studies on historical candidate genes and multi-omics data. Mol. Psychiatry 27, 95–112 (2022). 66. 66.Khandaker, G. M. & Dantzer, R. Is there a role for immune-to-brain communication in schizophrenia? Psychopharmacology (Berl). 233, 1559–1573 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00213-015-3975-1&link_type=DOI) 67. 67.Wu, Q. et al. Childhood and parental asthma, future risk of bipolar disorder and schizophrenia spectrum disorders: a population-based cohort study. Schizophr. Bull. 45, 360–368 (2019). 68. 68.Uellendahl-Werth, F. et al. Cross-tissue transcriptome-wide association studies identify susceptibility genes shared between schizophrenia and inflammatory bowel disease. *Commun*. Biol. 5, 80 (2022). 69. 69.Consortium, Gte. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science (80-.). 369, 1318–1330 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjkvNjUwOS8xMzE4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMTcvMjAyNC4wMi4xNi4yNDMwMjg4NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 70. 70.Shoubridge, A. P. et al. The gut microbiome and mental health: advances in research and emerging priorities. Mol. Psychiatry 27, 1908–1919 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41380-022-01479-w&link_type=DOI) 71. 71.Chauquet, S. et al. Association of Antihypertensive Drug Target Genes With Psychiatric Disorders: A Mendelian Randomization Study. JAMA psychiatry 78, 623– 631 (2021). 72. 72.Ryan, D. K. et al. Genetic Evidence for Protective Effects of Angiotensin-Converting Enzyme Against Alzheimer Disease But Not Other Neurodegenerative Diseases in European Populations. Neurol. Genet. 8, e200014 (2022). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoibm5nIjtzOjU6InJlc2lkIjtzOjExOiI4LzUvZTIwMDAxNCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAyLzE3LzIwMjQuMDIuMTYuMjQzMDI4ODUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 73. 73.Mayeda, E. R. et al. A simulation platform for quantifying survival bias: an application to research on determinants of cognitive decline. Am. J. Epidemiol. 184, 378–387 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwv451&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27578690&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 74. 74.Noyce, A. J. et al. Estimating the causal influence of body mass index on risk of Parkinson disease: a Mendelian randomisation study. PLoS Med. 14, e1002314 (2017). 75. 75.Michael Wyss, J., Kadish, I. & van Groen, T. AgelJRelated Decline in Spatial Learning and Memory: Attenuation by Captopril. Clin. Exp. Hypertens. 25, 455–474 (2003). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1081/CEH-120024988&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14596369&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185689500006&link_type=ISI) 76. 76.Mitchell, R. E. et al. Strategies to investigate and mitigate collider bias in genetic and Mendelian randomisation studies of disease progression. PLoS Genet. 19, e1010596 (2023). 77. 77.Owen, M. J., O’Donovan, M. C., Thapar, A. & Craddock, N. Neurodevelopmental hypothesis of schizophrenia. Br. J. Psychiatry 198, 173–175 (2011). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImJqcHJjcHN5Y2giO3M6NToicmVzaWQiO3M6OToiMTk4LzMvMTczIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMTcvMjAyNC4wMi4xNi4yNDMwMjg4NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 78. 78.Burgess, S., Davies, N. M. & Thompson, S. G. Bias due to participant overlap in two-sample Mendelian randomization. Genet. Epidemiol. 40, 597–608 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/gepi.21998&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27625185&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F02%2F17%2F2024.02.16.24302885.atom) 79. 79.Sadreev, I. I. et al. Navigating sample overlap, winner’s curse and weak instrument bias in Mendelian randomization studies using the UK Biobank. medRxiv 2021.06.28.21259622 (2021) doi:10.1101/2021.06.28.21259622. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wNi4yOC4yMTI1OTYyMnYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDIvMTcvMjAyNC4wMi4xNi4yNDMwMjg4NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=)