This article provides a comprehensive overview of trans-ancestry meta-analysis methodologies specifically applied to endometriosis genome-wide association studies (GWAS).
This article provides a comprehensive overview of trans-ancestry meta-analysis methodologies specifically applied to endometriosis genome-wide association studies (GWAS). Covering foundational principles to advanced applications, we explore how integrating diverse genetic datasets enhances discovery power, improves risk prediction, and reveals population-specific disease mechanisms. Key topics include novel computational frameworks for cross-ancestry integration, optimization strategies addressing genetic architecture heterogeneity, validation approaches for polygenic risk scores across populations, and therapeutic target identification through multi-omics integration. Designed for researchers, geneticists, and drug development professionals, this guide synthesizes cutting-edge methodologies from recent large-scale studies to advance precision medicine for endometriosis across global populations.
Endometriosis is a chronic, estrogen-driven inflammatory disorder affecting approximately 10% of reproductive-aged women globally, with diagnosis often delayed by 7-11 years from symptom onset [1] [2]. This application note examines the genetic architecture of endometriosis within the context of trans-ancestry meta-analysis methods, addressing how advanced genomic approaches are unraveling the disease's substantial heritable component. Twin and familial studies consistently demonstrate that endometriosis has a ~50% heritability rate, with approximately half of this genetic influence (26%) attributable to common single nucleotide polymorphisms (SNPs) [3]. Despite significant advances in genome-wide association studies (GWAS), which have identified 42 genomic loci associated with endometriosis risk, these common variants explain only ~5% of disease variance [2] [4], highlighting the need for more sophisticated analytical frameworks to capture the full genetic complexity.
Table 1: Key Heritability Estimates in Endometriosis
| Genetic Component | Estimate | Source Evidence | Notes |
|---|---|---|---|
| Overall Heritability | ~50% | Twin studies [3] | Proportion of disease risk in population due to genetic factors |
| Common SNP Contribution | ~26% | GWAS meta-analyses [3] | Proportion of heritability explained by common variants |
| GWAS-Explained Variance | ~5% | 42 identified loci [2] [4] | Current limitation of traditional GWAS approaches |
| Familial Risk Increase | 5-7 fold | First-degree relatives [3] | Compared to general population risk |
Beyond traditional GWAS findings, recent research has revealed several sophisticated layers of genetic complexity in endometriosis. Combinatorial genetics approaches have identified 1,709 disease signatures comprising 2,957 unique SNPs in combinations of 2-5 SNPs that significantly associate with endometriosis risk [2]. These multi-variant signatures explain substantially more disease risk than individual SNPs alone and highlight pathways involved in cell adhesion, proliferation, cytoskeleton remodeling, angiogenesis, fibrosis, and neuropathic pain [2] [4].
Integration of expression quantitative trait loci (eQTL) data from six physiologically relevant tissues (uterus, ovary, vagina, colon, ileum, and blood) has demonstrated tissue-specific regulatory effects of endometriosis-associated variants [5]. This tissue-specific regulation pattern suggests that genetic risk manifests differently across pelvic structures, with immune and epithelial signaling genes predominating in intestinal tissues, while reproductive tissues show enrichment of genes involved in hormonal response, tissue remodeling, and adhesion [5].
Interestingly, studies exploring ancient genetic contributions have identified regulatory variants derived from Neandertal and Denisovan introgression in genes including IL-6, CNR1, and IDO1 [1]. These ancient variants demonstrate significant enrichment in endometriosis cohorts and potentially interact with modern environmental pollutants, particularly endocrine-disrupting chemicals (EDCs), suggesting a novel evolutionary-environmental interplay in disease susceptibility [1].
The development of effective trans-ancestry meta-analysis methods faces significant challenges due to the variable genetic architecture of endometriosis across populations. Recent combinatorial analyses demonstrated that disease signatures identified in white European cohorts showed high reproducibility rates (80-88%) in the multi-ancestry All of Us cohort for high-frequency signatures (>9%), but substantially lower reproducibility (66-76%) for signatures with >4% frequency in non-white European sub-cohorts [2] [4]. This population-specificity underscores the critical need for diverse recruitment in genetic studies to ensure equitable advancement in endometriosis diagnosis and treatment across all ancestral backgrounds.
Table 2: Emerging Genetic Paradigms in Endometriosis Research
| Genetic Paradigm | Key Finding | Research Implications |
|---|---|---|
| Combinatorial Genetics | 75 novel genes identified beyond GWAS hits [2] [4] | Reveals complex multi-SNP interactions; identifies new biological pathways |
| Gene-Environment Interaction | Ancient variants interact with modern EDCs [1] | Suggests environmental triggers for genetically susceptible individuals |
| Tissue-Specific Regulation | Distinct eQTL effects across 6 relevant tissues [5] | Explains tissue-specific manifestation of lesions and symptoms |
| Cross-Ancestry Variation | Differential signature reproducibility [2] [4] | Highlights need for diverse cohorts in genetic studies |
| Pleiotropy with Comorbidities | Shared loci with pain conditions, osteoarthritis [3] | Explains comorbidity patterns and identifies shared therapeutic targets |
Traditional GWAS approaches examine single variant associations, limiting their ability to detect complex multi-variant interactions. Combinatorial analytics identifies combinations of 2-5 SNPs that collectively associate with endometriosis risk, revealing substantially more of the genetic architecture than conventional methods [2] [4].
Step 1: Cohort Selection and Quality Control
Step 2: Combinatorial Analysis
Step 3: Validation in Independent Cohorts
Step 4: Functional Annotation
Mendelian randomization uses genetic variants as instrumental variables to infer causal relationships between modifiable risk factors (e.g., metabolites, proteins) and endometriosis, reducing confounding bias inherent in observational studies [6].
Step 1: Instrumental Variable Selection
Step 2: Two-Sample Mendelian Randomization
Step 3: Colocalization Analysis
Step 4: Experimental Validation
Most endometriosis-associated GWAS variants reside in non-coding regions, suggesting they exert effects through gene regulation rather than protein coding changes. Mapping these variants to expression quantitative trait loci (eQTLs) across disease-relevant tissues reveals their regulatory consequences [5].
Step 1: Variant Curation and Annotation
Step 2: Multi-Tissue eQTL Mapping
Step 3: Tissue-Specific Functional Profiling
Step 4: Integration with Ancient Variation Data
Table 3: Essential Research Reagent Solutions for Endometriosis Genetic Studies
| Reagent/Resource | Function/Application | Example Sources/Platforms |
|---|---|---|
| PrecisionLife Combinatorial Analytics | Identifies multi-SNP disease signatures beyond GWAS | PrecisionLife Ltd. [2] [4] |
| GTEx v8 Database | Provides multi-tissue eQTL data for functional annotation | GTEx Portal [5] |
| SOMAscan Proteomics Platform | Measures 4,907 plasma protein levels for pQTL studies | SOMAscan V4 [6] |
| UK Biobank & All of Us Data | Large-scale genetic and health data for discovery/validation | UK Biobank, All of Us [2] [4] |
| Human R-Spondin3 ELISA Kit | Quantifies RSPO3 protein levels in validation studies | BOSTER Biological Technology [6] |
| Ensembl VEP | Functional annotation of genetic variants | Ensembl [1] [5] |
| LDlink Suite | Linkage disequilibrium and population genetics analysis | LDlink, LDpop, LDpair [1] |
| MSigDB Hallmark Gene Sets | Functional enrichment analysis for biological interpretation | Molecular Signatures Database [5] |
| 4,5-Dibromooctane | 4,5-Dibromooctane|CAS 61539-75-1|Supplier | 4,5-Dibromooctane is a vicinal dibromide for organic synthesis research. For Research Use Only. Not for human or veterinary use. |
| Gnidilatin | Gnidilatin, CAS:60195-69-9, MF:C37H48O10, MW:652.8 g/mol | Chemical Reagent |
The integration of trans-ancestry meta-analysis with combinatorial genetics, Mendelian randomization, and functional genomic approaches represents a powerful framework for elucidating the complex genetic architecture of endometriosis. These advanced methods have already identified 75 novel gene associations beyond traditional GWAS findings [2], revealed causal relationships with specific plasma proteins like RSPO3 [6], and demonstrated how ancient regulatory variants interact with modern environmental factors to influence disease risk [1]. As these approaches are refined and applied to increasingly diverse populations, they promise to accelerate the development of improved diagnostic biomarkers, personalized risk prediction tools, and novel therapeutic strategies for this complex and debilitating condition.
Endometriosis, a chronic inflammatory condition affecting approximately 10% of reproductive-aged women globally, demonstrates substantial genetic susceptibility with heritability estimates reaching 47% [1]. While genome-wide association studies (GWAS) have identified numerous susceptibility loci, the predominant reliance on European-ancestry cohorts has created significant blind spots in our understanding of the disease's genetic architecture across diverse populations. This application note examines the methodological limitations of European-centric GWAS approaches and outlines trans-ancestry meta-analysis protocols to advance more inclusive genetic research in endometriosis.
Table 1: Documented Limitations of European-Centric GWAS in Endometriosis Research
| Limitation Category | Specific Challenge | Documented Evidence |
|---|---|---|
| Population-Specific Alleles | Risk alleles identified in European populations show different effects in other ancestries | Sardinian population study showed no significant association for variants significant in other European groups [7] |
| Variant Spectrum | Limited capture of ancestry-specific genetic variations | Iranian population study identified unique SNP associations in MFN2, PINK1, and PRKN genes [7] |
| Regulatory Complexity | Tissue-specific eQTL effects not fully characterized across ancestries | Multi-tissue eQTL analysis revealed tissue-specific regulatory profiles for endometriosis risk variants [8] [5] |
| Gene-Environment Interactions | Incomplete understanding of how genetic risks interact with diverse environmental exposures | Ancient regulatory variants from Neandertal introgression show potential interaction with modern environmental pollutants [1] |
The fundamental assumption that genetic discoveries in European populations readily translate to other ancestries has repeatedly proven problematic in endometriosis research. Studies across diverse populations have demonstrated differential effect sizes and heterogeneous genetic architecture. In the Sardinian population, for instance, variants significantly associated with endometriosis in other European cohorts showed no significant association, suggesting that specific risk alleles could act differently in the pathogenesis of the disease across ethnic populations [7]. Similarly, research in Iranian women identified unique single nucleotide polymorphism (SNP) associations in genes involved in mitophagy (MFN2, PINK1, and PRKN) that were not highlighted in major European GWAS [7].
European-centric approaches have insufficiently characterized the tissue-specific regulatory landscape of endometriosis risk variants across diverse ancestries. A comprehensive multi-tissue eQTL analysis demonstrated that endometriosis-associated variants exhibit distinct regulatory profiles across six physiologically relevant tissues (uterus, ovary, vagina, sigmoid colon, ileum, and peripheral blood) [8] [5]. In reproductive tissues, these variants preferentially regulated genes involved in hormonal response, tissue remodeling, and adhesion, while in intestinal tissues and blood, they predominantly affected immune and epithelial signaling genes [5]. This tissue specificity underscores how limited ancestral diversity in GWAS reduces our ability to identify the full spectrum of regulatory mechanisms contributing to endometriosis pathogenesis.
European-centric GWAS designs have historically struggled to account for the diverse environmental exposures that interact with genetic risk factors across global populations. Emerging evidence suggests that ancient regulatory variants, some originating from Neandertal introgression, may interact with modern environmental pollutants to modulate endometriosis risk [1]. Co-localized IL-6 variants (rs2069840 and rs34880821) located at a Neandertal-derived methylation site demonstrated strong linkage disequilibrium and potential immune dysregulation in response to contemporary environmental triggers [1]. The restricted ancestral diversity in most GWAS limits statistical power to detect such gene-environment interactions, which likely vary substantially across populations with different historical evolutionary pressures and modern environmental exposures.
The trans-ancestry meta-analysis protocol provides a robust methodological framework to overcome limitations of European-centric GWAS. This approach integrates diverse ancestry groups while accounting for population-specific genetic architectures, as demonstrated in recent large-scale fibroid research that included 74,294 cases (27.7% non-European descent) and 465,810 controls (18.3% non-European descent) [9].
Table 2: Trans-Ancestry Meta-Analysis Protocol for Endometriosis Genomics
| Protocol Stage | Key Procedures | Ancestry-Specific Considerations |
|---|---|---|
| Cohort Selection | Identify diverse biobanks and study populations | Ensure representative sampling across ancestry groups with careful population stratification control |
| Quality Control | Implement standardized SNP filtering and imputation | Apply ancestry-specific reference panels for imputation; account for differential allele frequencies |
| Association Testing | Perform ancestry-stratified GWAS followed by meta-analysis | Use ancestry-appropriate linkage disequilibrium reference panels; apply genomic control inflation factors |
| Heritability Estimation | Calculate SNP-based heritability within and across ancestries | Utilize ancestry-specific HapMap3 annotated tags; compare heritability estimates across groups |
| Functional Annotation | Integrate multi-omics data for putative causal gene identification | Incorporate ancestry-specific eQTL/pQTL maps when available; account for tissue-specific regulation |
Experimental Protocol 1: Trans-Ancestry GWAS Meta-Analysis
Cohort Acquisition and Harmonization
Ancestry-Stratified Analysis
Cross-Ancestry Meta-Analysis
Variant Prioritization and Validation
The integrative multi-omic protocol leverages Mendelian randomization and colocalization approaches to bridge the gap between genetic associations and functional mechanisms across diverse populations, addressing a critical limitation of European-centric studies that often prioritize coding variants over regulatory elements.
Experimental Protocol 2: Multi-Omic Integration for Cross-Ancestry Functional Validation
Multi-Omic Data Acquisition
Summary-based Mendelian Randomization (SMR) Analysis
Colocalization Analysis
Cross-Ancestry Functional Validation
Table 3: Key Research Reagents for Trans-Ancestry Endometriosis Genomics
| Reagent/Category | Specific Examples | Application in Endometriosis Research |
|---|---|---|
| Genotyping Arrays | Illumina Global Screening Array, UK Biobank Axiom Array | Genome-wide variant detection in diverse populations with ancestry-informative markers |
| eQTL Resources | GTEx v8 database, eQTLGen consortium | Tissue-specific expression quantitative trait loci mapping across multiple tissues relevant to endometriosis |
| pQTL Platforms | SOMAscan V4 assay (4,907 cis-pQTLs) [6] | Plasma protein quantitative trait loci identification for therapeutic target prioritization |
| Methylation Analysis | Illumina Infinium MethylationEPIC array | Genome-wide DNA methylation profiling to identify epigenetic regulators of endometriosis risk |
| Validation Assays | Human R-Spondin3 ELISA Kit [6], TRIzol reagent for RNA extraction [7] | Experimental validation of candidate biomarkers and therapeutic targets in clinical samples |
| Bioinformatics Tools | SMR software v1.3.1, COLOC R package, LDlink [10] | Statistical analysis of multi-omic data integration and colocalization evidence |
| Gardmultine | Gardmultine | Gardmultine is a bis-indole alkaloid for research, studied for its antitumor properties and complex spirocyclic structure. For Research Use Only. Not for human use. |
| 3-Methyldiaziridine | 3-Methyldiaziridine|C4H10N2|RUO | 3-Methyldiaziridine (CAS 4901-75-1) is a valuable reagent for chemical research. This product is For Research Use Only and is not intended for personal use. |
The historical limitations of European-centric GWAS in endometriosis research have created significant gaps in our understanding of the disease's genetic architecture across global populations. The implementation of trans-ancestry meta-analysis protocols, coupled with integrative multi-omic approaches, provides a robust framework to overcome these limitations. By embracing methodological innovations that prioritize ancestral diversity, the research community can accelerate the discovery of novel therapeutic targets like RSPO3 [6] [11] and develop more effective, personalized interventions for endometriosis across all populations. Future directions should include expanded recruitment from underrepresented ancestries, development of ancestry-specific reference panels, and dedicated funding initiatives to support diverse cohort collection and analysis.
Large-scale genetic studies have fundamentally advanced our understanding of endometriosis pathophysiology, moving beyond association to reveal causative mechanisms. Trans-ancestry meta-analyses of genome-wide association studies (GWAS) have been particularly instrumental, identifying risk loci across diverse populations and enabling a more precise dissection of the molecular basis of the disease [12] [13]. These studies consistently demonstrate that genetic susceptibility converges on a limited set of core biological pathways. This application note synthesizes the latest genetic and multi-omic evidence to detail three principal pathwaysâhormone signaling, immune regulation, and tissue remodelingâand provides standardized protocols for their investigation in functional studies. By framing these insights within modern genomic methodologies, we aim to equip researchers with the tools necessary to translate genetic discoveries into targeted therapeutic strategies.
Integrative analysis of GWAS data with expression quantitative trait loci (eQTLs), methylation QTLs (mQTLs), and protein QTLs (pQTLs) has illuminated the functional impact of non-coding risk variants, revealing their tissue-specific regulatory effects and their convergence on key pathogenic processes [8] [10]. The table below summarizes the core pathways, key genetic findings, and implicated cell types.
Table 1: Key Biological Pathways and Genetic Findings in Endometriosis
| Biological Pathway | Key Genes/Proteins from Genetic Studies | Primary Functions & Mechanisms | Relevant Tissues/Cell Types |
|---|---|---|---|
| Hormone Signaling | WNT4, GREB1, FSHB, ESR1, RSPO3 |
Regulation of estrogen-responsive genes; Müllerian duct development; estrogen-driven proliferation [14] [15]. | Ovary, Uterus, Endometriotic lesions |
| Immune Regulation | MICB, IL-6, IDO1, CNR1 |
Immune evasion; chronic inflammation; altered T-cell function; pain sensitization [8] [1]. | Peripheral blood, Intestinal tissues, Lesions |
| Tissue Remodeling & Cell Adhesion | VEZT, FN1, MAP3K5, ENG, FLT1 |
Ectopic tissue anchoring; cell survival; apoptosis resistance; angiogenesis [10] [6] [14]. | Uterus, Sigmoid colon, Ileum, Lesions |
The hormone signaling pathway is central to endometriosis, an estrogen-dependent disease. Genetic studies have robustly identified loci within genes that are critical for reproductive tract development and estrogen-mediated proliferation.
WNT4 is a consistently replicated risk locus. It is crucial for Müllerian duct development and functions as a key regulator of steroid hormone action in the endometrium. Trans-ancestry GWAS have identified an intronic variant in WNT4 (rs61768001) associated with multiple subtypes of female infertility, which is a common comorbidity of endometriosis [16] [15]. Similarly, GREB1 is an estrogen-responsive gene implicated in cell cycle control, and FSHB is involved in gonadotropin regulation [14] [15].Immune dysregulation is a hallmark of endometriosis, and genetic studies pinpoint a role for both systemic and local immune dysfunction in disease susceptibility.
MICB (a stress-induced ligand for immune cells) is consistently linked to immune evasion pathways in peripheral blood and intestinal tissues [8]. Additionally, regulatory variants in IL-6 (a pro-inflammatory cytokine), some of which are linked to a Neandertal-derived methylation site, demonstrate significant enrichment in endometriosis cohorts and are implicated in immune dysregulation [1].IL-6 and IDO1 variants may skew immune responses, while CNR1 (the cannabinoid receptor 1 gene) variants also suggest a genetic link to pain sensitization, a core symptom of the disease [1].The ability of ectopic endometrial tissue to implant, invade, and persist requires significant remodeling of the extracellular matrix and establishment of a new blood supply.
VEZT encodes a cell adhesion protein that may facilitate the anchoring of ectopic tissue [15]. Multi-omic SMR analyses have identified MAP3K5 as a key gene, where specific methylation patterns causally downregulate its expression and heighten endometriosis risk, potentially by affecting cell survival and stress responses [10]. Proteomic MR studies have also implicated FLT1 (a VEGF receptor) and ENG (Endoglin) in disease risk, underscoring the role of angiogenesis [10] [6].MAP3K5, a gene involved in stress-induced apoptosis, may confer resistance to cell death in ectopic lesions, while FLT1 and ENG drive the vascularization necessary for lesion growth and maintenance [10] [6].
Diagram Title: From Genetic Loci to Disease Pathogenesis
Objective: To determine if endometriosis-associated genetic variants regulate gene expression in a tissue-specific manner.
Background: Most GWAS-identified variants reside in non-coding regions. eQTL analysis tests their association with gene expression levels, providing a functional link between genetics and pathophysiology [8].
Materials:
Procedure:
Expected Output: A list of genes whose expression is significantly regulated by endometriosis risk variants in each analyzed tissue, highlighting genes like MICB in blood or WNT4 in the uterus [8].
Objective: To investigate the causal effect of a mediating molecular trait (gene expression, DNA methylation, protein abundance) on endometriosis risk.
Background: SMR integrates GWAS summary data with QTL data to test if variation in a molecular phenotype is causally associated with the disease [10].
Materials:
Procedure:
Expected Output: Identification of putatively causal genes and proteins (e.g., MAP3K5, ENG) whose altered regulation, driven by genetic variation, influences endometriosis risk [10].
Table 2: Key Research Reagent Solutions for Endometriosis Pathway Analysis
| Reagent / Resource | Function / Application | Example Source / Catalog |
|---|---|---|
| GTEx v8 Database | Reference dataset for tissue-specific eQTL analysis. | GTEx Portal ( [8]) |
| SOMAscan Platform | Multiplexed proteomic assay for pQTL discovery and validation. | SomaLogic ( [6]) |
| coloc R Package | Bayesian test for colocalization between GWAS and QTL signals. | CRAN ( [10]) |
| SMR Software | Tool for multi-omic Summary-based Mendelian Randomization analysis. | CNS Genomics ( [10]) |
| Human R-Spondin3 ELISA Kit | Quantitative measurement of RSPO3 protein levels in plasma or tissue. | BOSTER Biological Technology ( [6]) |
Diagram Title: Integrative Genomics Workflow for Target Discovery
The integration of large-scale trans-ancestry GWAS with multi-omic data has definitively established that genetic risk for endometriosis is channeled through dysregulation in hormone signaling, immune function, and tissue remodeling. The application of standardized protocols for eQTL and SMR analysis, as detailed herein, provides a robust framework for the scientific community to move from genetic associations to a mechanistic understanding of disease. The continued growth of diverse, large-scale biobanks, coupled with the functional tools in the Scientist's Toolkit, will be critical for translating these key biological pathways into much-needed diagnostic and therapeutic advancements for endometriosis.
Endometriosis demonstrates significant variation in its epidemiological presentation across different ancestral groups. Recent large-scale meta-analyses provide comprehensive quantitative assessments of this heterogeneity, essential for guiding trans-ancestry genetic research and clinical drug development strategies.
Table 1: Global Prevalence of Endometriosis Across Populations and Clinical Subtypes
| Population / Subtype | Prevalence (%) | 95% Confidence Interval | Data Source |
|---|---|---|---|
| General Population | 5 | 2-9 | [17] |
| Women with Infertility | 38 | 25-51 | [17] |
| Symptomatic Women | 18-42 | Not specified | [17] |
| Peritoneal Endometriosis | 6 | 1-15 | [17] |
| Ovarian Endometriosis | 13 | 5-24 | [17] |
| Deep Endometriosis | 10 | 2-24 | [17] |
| Nonane-2,5-diol | Nonane-2,5-diol, CAS:51916-45-1, MF:C9H20O2, MW:160.25 g/mol | Chemical Reagent | Bench Chemicals |
| Dec-9-yn-4-ol | Dec-9-yn-4-ol|C10H18O|Research Chemical | High-purity Dec-9-yn-4-ol for research applications. This product is For Research Use Only. Not for human or veterinary use. | Bench Chemicals |
The global prevalence of endometriosis is estimated at approximately 5% in the general population, rising dramatically to 38% among women experiencing infertility [17]. When examining disease subtypes, ovarian endometriosis represents the most common presentation at 13%, followed by deep endometriosis (10%) and peritoneal endometriosis (6%) [17]. These differential prevalence rates across clinical manifestations highlight the disease's substantial heterogeneity.
Geographical and ancestral analyses reveal a nine-fold increased risk among women of East Asian ancestry compared to European or American populations [18]. This disparity underscores the critical importance of accounting for ancestral background in both genetic research and therapeutic development.
Genome-wide association studies (GWAS) have identified numerous susceptibility loci for endometriosis, with effect sizes and prevalence varying significantly across ancestral groups.
Table 2: Key Endometriosis Genetic Loci and Ancestral Heterogeneity
| Genetic Locus | Nearest Gene | Reported Function | Ancestral Heterogeneity |
|---|---|---|---|
| rs7521902 | WNT4 | Sex steroid hormone signaling | Stronger association in European ancestry |
| rs10965235 | CDKN2B-AS1 | Cell cycle regulation | Initially identified in Japanese ancestry [19] |
| rs12700667 | Intergenic 7p15.2 | Developmental pathways | Consistent across populations [19] [20] |
| rs13394619 | GREB1 | Hormone-mediated growth | Stronger effect in Stage III/IV disease [19] [20] |
| rs1250248 | FN1 | Sex steroid hormone pathways | Associated with moderate-severe disease [21] [20] |
Recent multi-ancestry genetic research has substantially expanded our understanding of endometriosis risk loci. A 2024 Mendelian randomization study incorporating trans-ethnic analyses confirmed consistent directions of effect for seven out of nine established loci across European and East Asian populations [22]. The most recent and largest multi-ancestry GWAS to date (2025), comprising approximately 1.4 million women (105,869 cases), identified 80 genome-wide significant associations, 37 of which are novel [12]. This study provided the first genetic variants specifically associated with adenomyosis, a related gynecological condition [12].
Population genomic analyses examining disease genomic 'grammar' have identified 296 common genetic targets with low allele frequencies and 6 with high allele frequencies across five major population groups (Europeans, Africans, Americans, East Asians, and South Asians) [18]. The substantial variation in genetic architecture observed across populations reflects both divergent evolutionary histories and environmental interactions.
Objective: To establish a standardized protocol for trans-ancestry meta-analysis of endometriosis genome-wide association studies, enabling the identification of genetic risk factors across diverse populations.
Inclusion Criteria:
Stratification Approach:
Sample Size Requirements: Minimum 5,000 cases per ancestral group for adequate statistical power in trans-ancestry analyses [12]
Genotyping Platforms: Utilize high-density GWAS arrays (e.g., Illumina Global Screening Array, Affymetrix Axiom Biobank Array)
Quality Control Steps:
Imputation Protocol:
Primary Association Analysis:
Meta-Analysis Approach:
Conditional Analysis:
Functional Annotation:
Trans-ancestry GWAS workflow illustrating the iterative process for identifying heterogeneous genetic effects across populations.
Genetic studies consistently implicate genes involved in sex steroid hormone biosynthesis and signaling in endometriosis pathogenesis. Key pathways identified through trans-ancestry analyses include:
Estrogen Receptor Signaling:
Progesterone Resistance Pathways:
Endometriosis pathogenesis network showing how genetic risk variants influence multiple biological pathways contributing to clinical heterogeneity.
Emerging evidence from Mendelian randomization studies indicates causal relationships between lipid metabolism and endometriosis risk:
Table 3: Essential Research Reagents for Endometriosis Trans-ancestry Studies
| Reagent Category | Specific Examples | Research Application | Protocol Considerations |
|---|---|---|---|
| Genotyping Arrays | Illumina Global Screening Array, Affymetrix Axiom Biobank Array | Genome-wide variant detection | Ancestry-specific content optimization [18] |
| Imputation Panels | 1000 Genomes Phase 3, TOPMed, HRC | Genotype gap filling | Multi-ancestry reference panels improve imputation accuracy [18] |
| Functional Validation | CRISPR/Cas9 systems, organoid culture models | Mechanism investigation | Patient-derived organoids from diverse ancestries [12] |
| Bioinformatics Tools | METAL, REGENIE, GCTA, PLINK | Statistical analysis | Trans-ancestry meta-analysis software [19] [20] |
| Pathway Analysis | GARFIELD, FUMA, DEPICT | Functional annotation | Integration with multi-omic databases [12] |
| Dehydrobruceantarin | Dehydrobruceantarin - CAS 53663-00-6 | Dehydrobruceantarin is a natural product for research. This product is For Research Use Only. Not for human or veterinary diagnostic or therapeutic use. | Bench Chemicals |
| Butanoyl azide | Butanoyl Azide|C4H7N3O|Research Chemical | Butanoyl azide for research applications. This compound is For Research Use Only. Not for diagnostic, therapeutic, or personal use. | Bench Chemicals |
Step 1: Chromatin State Mapping
Step 2: Colocalization Analysis
Step 3: High-Throughput Functional Screens
Cell Culture Models:
Functional Assays:
This integrated framework for trans-ancestry analysis of endometriosis provides a comprehensive approach to elucidate the genetic architecture and biological mechanisms underlying this complex gynecological disorder, with direct implications for targeted therapeutic development across diverse populations.
Recent advances in trans-ancestry genomic research have dramatically accelerated the discovery of genetic loci associated with endometriosis. This application note details how multi-ancestry genome-wide association studies (GWAS) have expanded the catalog of significant endometriosis loci from approximately 45 to over 80 through the inclusion of diverse cohorts. We present quantitative evidence from a landmark study of ~1.4 million women, including 105,869 cases, which identified 80 genome-wide significant associationsâ37 of which are novel. This document provides detailed methodologies for implementing trans-ancestry meta-analysis approaches, including specific protocols for statistical analysis, functional annotation, and therapeutic target discovery. The presented framework demonstrates how genetic studies encompassing diverse ancestral backgrounds enhance discovery power, improve fine-mapping resolution, and facilitate the translation of genetic findings into pathogenic mechanisms and potential therapeutic interventions.
Endometriosis is a common, estrogen-dependent inflammatory condition affecting approximately 10% of women of reproductive age, characterized by the presence of endometrial-like tissue outside the uterine cavity [8]. The disease presents with chronic pelvic pain, dysmenorrhea, and infertility, with significant impacts on quality of life. The heritability of endometriosis is estimated at around 52%, highlighting the substantial role of genetic factors in disease pathogenesis [19].
Early GWAS efforts in endometriosis identified approximately 45 significant genetic loci, primarily in populations of European and Japanese ancestry [19]. However, these studies were limited by their predominantly single-ancestry focus, which constrained discovery power and fine-mapping resolution. The recent application of trans-ancestry meta-analysis approaches has dramatically expanded our understanding of the genetic architecture of endometriosis, increasing the number of genome-wide significant loci to over 80 [12] [13].
This application note documents the methodologies and protocols that enabled this expansion, focusing specifically on the integration of diverse cohorts in endometriosis genetics research. We present comprehensive data from a recent multi-ancestry GWAS of ~1.4 million women, experimental protocols for trans-ancestry meta-analysis, and visualization of key signaling pathways implicated by the discovered loci.
Table 1: Chronological Expansion of Significant Endometriosis Loci
| Study Period | Sample Size | Cases | Significant Loci | Key Genetic Findings | Population Focus |
|---|---|---|---|---|---|
| Pre-2023 Meta-analyses | ~44,000 | ~11,500 | ~45 | Associations near WNT4, VEZT, GREB1 | Primarily European and Japanese |
| 2023 Nature Genetics | 762,601 | 60,674 | 42 (49 signals) | Shared pathways with pain conditions | European and East Asian |
| 2025 Multi-ancestry GWAS | ~1.4 million | 105,869 | 80 (37 novel) | First adenomyosis loci; immune and tissue remodeling pathways | Multi-ancestry |
Table 2: Cohort Characteristics in Landmark Endometriosis GWAS
| Ancestral Group | 2023 Study (N=762,601) | 2025 Study (N=~1.4M) | Notable Population-Specific Findings |
|---|---|---|---|
| European | Primary focus | Expanded inclusion | Strongest associations with stage III/IV disease |
| East Asian | Included | Included | Consistent effect directions with European associations |
| African | Limited representation | Increased inclusion | Improved fine-mapping resolution |
| Other Ancestries | Limited | Expanded | Novel loci discovery in admixed populations |
The 2025 multi-ancestry GWAS represents the largest genetic study of endometriosis to date, achieving a 78% increase in significant loci compared to pre-2023 findings [12] [13]. This expansion was facilitated by a 318% increase in sample size and deliberate inclusion of diverse ancestral groups, enabling the discovery of 37 novel loci and the first five genetic variants associated with adenomyosis [13].
Purpose: To detect and fine-map complex trait association signals while accounting for heterogeneity in allelic effects correlated with ancestry.
Materials:
Procedure:
filelist.txt contains paths to all GWAS summary statistics files.Validation: Compare power and fine-mapping resolution against fixed-effects and random-effects meta-analysis using simulated datasets [24].
Purpose: To characterize the functional consequences of identified variants through transcriptomic, epigenetic, and proteomic data integration.
Materials:
Procedure:
Validation: Prioritize candidate genes based on (1) number of associated eQTL variants and (2) magnitude of regulatory effect (slope value) across multiple tissues [8].
The biological pathways illuminated by the expanded genetic discoveries reveal a complex interplay of mechanisms in endometriosis pathogenesis. Multi-omics integration demonstrates that genetic variation influences disease risk through transcriptomic, epigenetic, and proteomic regulation across multiple tissues, converging on pathways involved in immune regulation, tissue remodeling, and cell differentiation [12] [13]. Specific genes identified in these pathways include:
Notably, a substantial subset of regulated genes was not associated with any known pathway, indicating potential novel regulatory mechanisms in endometriosis pathogenesis [8].
Table 3: Essential Research Materials for Endometriosis Genetic Studies
| Reagent/Resource | Specific Example | Function/Application | Key Features |
|---|---|---|---|
| GWAS Arrays | Illumina Global Screening Array | Genotyping of common variants | ~650,000 markers with imputation to millions of variants |
| Reference Panels | 1000 Genomes Phase 3 | Imputation and ancestry determination | 2,504 individuals from 26 populations |
| eQTL Databases | GTEx v8 | Tissue-specific expression quantitative trait loci | 54 tissues from 948 donors |
| Meta-analysis Software | MR-MEGA | Trans-ethnic meta-regression | Accounts for ancestry-correlated heterogeneity |
| Functional Annotation | Ensembl VEP | Variant effect prediction | Genomic context and functional consequences |
| Pathway Analysis | MSigDB Hallmark | Biological pathway enrichment | 50 well-defined biological states |
| Colocalization Tools | coloc R package | Bayesian colocalization of GWAS and molecular QTLs | Determines shared causal variants |
| Cobalt;samarium | Cobalt;samarium (SmCo) Alloy | Cobalt;samarium (SmCo) magnet alloy for research applications in aerospace, electronics, and renewable energy. For Research Use Only (RUO). Not for personal use. | Bench Chemicals |
| Nickel-Wolfram | Nickel-Wolfram (Ni/W) Research Material | Bench Chemicals |
The expansion of endometriosis loci has enabled drug-repurposing analyses that highlight potential therapeutic interventions currently used for breast cancer and preterm birth prevention [12] [13]. These analyses leverage the integration of genetic findings with drug target databases to identify existing medications that might be effective for endometriosis treatment based on shared pathogenic mechanisms.
Genetic correlations between endometriosis and other pain conditions, including migraine, back pain, and multisite chronic pain (MCP), suggest that targeted investigations of shared mechanisms could aid the development of new treatments and facilitate early symptomatic intervention [25]. The polygenic risk for endometriosis has been shown to interact with abdominal pain, anxiety, migraine, and nausea, providing insights for managing the complex symptomatology of the condition [12].
The strategic inclusion of diverse cohorts in endometriosis genetic research has substantially expanded our understanding of the genetic architecture of this complex condition. The application of trans-ancestry meta-analysis methods has enabled the discovery of 37 novel loci in addition to the previously known 45 associations, providing a more comprehensive picture of the biological pathways involved in disease pathogenesis.
The protocols and methodologies detailed in this application note provide a roadmap for researchers seeking to implement similar approaches for other complex traits. The continued expansion of diverse biobanks and advancements in trans-ancestry analytical methods will further accelerate gene discovery, fine-mapping precision, and the translation of genetic findings into clinically actionable insights for endometriosis diagnosis and treatment.
Endometriosis is a heritable, hormone-dependent gynecological disorder affecting 6-10% of women of reproductive age, with an estimated common SNP-based heritability of 0.26 [21]. Trans-ancestry meta-analysis has emerged as a powerful approach for elucidating the genetic architecture of endometriosis, enabling the identification of susceptibility loci across diverse populations. Genome-wide association studies (GWAS) have successfully identified multiple genetic loci associated with endometriosis risk, with recent large-scale meta-analyses identifying five novel loci in genes involved in sex steroid hormone pathways (FN1, CCDC170, ESR1, SYNE1, and FSHB) [21]. However, the analysis of complex genetic data from diverse ancestral backgrounds presents significant methodological challenges, requiring advanced statistical frameworks to maximize discovery while ensuring equitable performance across populations.
The integration of polygenic risk scores (PRS), Bayesian modeling, and pathway-based approaches represents a paradigm shift in endometriosis genetics. These methods address critical limitations of conventional GWAS by incorporating SNPs with modest effect sizes, accounting for linkage disequilibrium differences across populations, and enabling multi-locus testing. This article outlines core analytical frameworks specifically applied to endometriosis research, providing detailed protocols for implementing PRS-CSx, Bayesian graphical models, and Adaptive Rank Truncated Product methods in trans-ancestry meta-analysis contexts.
Polygenic Risk Score (PRS) analysis predicts an individual's genetic risk for targeted traits by aggregating the effects of numerous genetic variants across the genome. Unlike conventional GWAS that focuses on statistically significant markers, PRS incorporates single nucleotide polymorphisms (SNPs) with low effect sizes that collectively contribute to disease heritability [26]. This approach is particularly valuable for endometriosis research, where the condition exhibits a complex polygenic architecture with contributions from many genetic variants of small effect.
The clinical application of PRS in endometriosis faces the significant challenge of reduced predictive power in non-European populations due to insufficient GWAS data and differences in genetic architecture [26]. A 2022 study investigating the applicability of PRS in endometriosis clinical presentation found inverse associations between PRS and spread of endometriosis, involvement of the gastrointestinal tract, and hormone treatment, though the specificity and sensitivity were low [27]. The authors concluded that specific PRS should be developed to predict clinical presentations in patients with endometriosis, highlighting the need for more sophisticated cross-population methods.
PRS-CSx is a Bayesian regression framework that addresses cross-population PRS applications by using a continuous shrinkage (CS) prior on SNP effect sizes and leveraging multi-ancestry reference panels [26]. This method improves the accuracy of PRS application across multi-ethnic populations through a posterior inference algorithm that accounts for genetic architecture differences between populations.
Table 1: Key PRS Methods and Their Applications in Endometriosis Research
| Method | Approach | Computational Platform | Key Features | Endometriosis Application |
|---|---|---|---|---|
| PRS-CSx | Bayesian shrinkage with continuous prior | Python | Multi-ancestry inference, improves cross-population portability | Trans-ancestry risk prediction for diverse cohorts |
| LDpred | Bayesian shrinkage prior | Python/R | Uses prior on effect sizes and LD information | Disease risk prediction in European populations |
| PRSice | Clumping + thresholding (C+T) | R, C++ | User-friendly, automated PRS analysis | Clinical presentation association studies |
| BayesR | Hierarchical Bayesian mixture model | Fortran | Simultaneous variant discovery and variance estimation | Modeling polygenic architecture |
Required Input Data:
Step-by-Step Procedure:
Data Preparation and Quality Control
LD Reference Panel Processing
PRS-CSx Execution
python PRScsx.py --sumstats1 EUR.txt --sumstats2 EAS.txt --ref1 EUR_ref --ref2 EAS_ref --out endometriosis_prsScore Calculation and Validation
plink --score endometriosis_prs.txt
Figure 1: PRS-CSx Workflow for Trans-ancestry Endometriosis Risk Prediction
Bayesian graphical models provide a powerful framework for multi-SNP analysis of GWAS data, addressing limitations of standard single-marker approaches. These methods enable simultaneous assessment of multiple SNPs that can be linked or unlinked and can interact or not, providing a more comprehensive understanding of genetic architecture [28]. For endometriosis research, this approach is particularly valuable given the complex, polygenic nature of the disease.
The fundamental advantage of Bayesian methods lies in their ability to model complex dependency structures among genetic variants while accounting for population structure and multiple testing through posterior probabilities [28]. Unlike single-SNP GWAS that test each marker independently, Bayesian graphical models evaluate the joint effect of multiple SNPs, potentially identifying combinations of variants that collectively influence endometriosis risk.
The "Bayesian Alphabet" encompasses a family of methods for genomic prediction and GWAS, each employing different prior distributions for marker effects [29]. Key methods include:
These methods have been shown to map quantitative trait loci (QTL) more precisely than standard single-SNP GWAS, with applications demonstrating higher accuracy for QTL detection in complex traits [29]. For endometriosis, which involves multiple genetic variants of small to moderate effects, Bayesian methods offer enhanced power to detect genuine associations.
Table 2: Bayesian Methods for Genomic Analysis in Endometriosis Research
| Method | Prior Distribution | Key Features | Implementation | Endometriosis Relevance |
|---|---|---|---|---|
| Bayes-A | Normal with marker-specific variance | Accommodates large effects | BGLR, Gensel | Captures effect size heterogeneity |
| Bayes-B | Mixture with point mass at zero | Variable selection capability | JWAS, BGLR | Identifies causal SNPs among thousands |
| Bayes-C | Single normal for non-zero effects | Intermediate complexity | Gensel, BGLR | Balanced approach for polygenic traits |
| Bayes-R | Mixture of normals | Models effect size distribution | BayesR software | Optimal for highly polygenic architecture |
Required Input Data:
Step-by-Step Procedure:
Data Preprocessing
Model Specification
Stochastic Search Execution
Posterior Inference
Figure 2: Bayesian Graphical Model Workflow for Endometriosis GWAS
The Adaptive Rank Truncated Product (ARTP) method provides a powerful approach for pathway-based meta-analysis using summary statistics from GWAS. This method enables multi-marker testing procedures that integrate information across multiple genetic variants within biological pathways, offering enhanced power to detect subtle polygenic effects [30] [31]. For endometriosis research, pathway analysis is particularly valuable given the involvement of multiple biological processes, including sex steroid hormone signaling and immune function.
The ARTP2 method, an enhanced version of the original algorithm, allows for association testing on user-defined genes or pathways without assuming independence between genes, making it suitable for analyzing overlapping functional pathways [30]. This approach can leverage summary statistics from trans-ancestry meta-analyses, facilitating the identification of biological pathways enriched for endometriosis risk variants across diverse populations.
The summary-based Adaptive Rank Truncated Product (sARTP) method enables pathway meta-analysis using only SNP-level summary statistics in combination with genotype correlation estimated from a reference panel [31]. This approach has been validated through comprehensive applications, including a pathway-based meta-analysis of type 2 diabetes that identified 43 significant pathways, demonstrating its utility for complex disease genetics.
For endometriosis research, sARTP enables the integration of summary statistics from multiple ancestries to identify conserved biological pathways, even when individual variant effects are heterogeneous across populations. This method is particularly valuable for trans-ancestry analysis where individual-level genotype data may not be available for all cohorts.
Required Input Data:
Step-by-Step Procedure:
Pathway Definition and Annotation
Summary Statistics Processing
ARTP2 Execution
Rscript ARTP2.R --sumstats endometriosis.txt --pathway hormone_pathways.txt --out pathway_resultsResults Interpretation
Figure 3: ARTP2 Pathway Analysis Workflow for Endometriosis Genetics
The integration of PRS-CSx, Bayesian modeling, and ARTP2 methods creates a powerful analytical framework for trans-ancestry endometriosis research. These approaches address complementary aspects of genetic analysis: PRS-CSx enables cross-population risk prediction, Bayesian methods identify multi-SNP associations, and ARTP2 elucidates biological pathways. When applied synergistically, these methods provide a comprehensive understanding of endometriosis genetics across diverse populations.
A recommended analytical sequence begins with trans-ancestry meta-analysis to identify robust genetic associations, followed by Bayesian graphical modeling to refine multi-SNP models, then pathway analysis to identify biological mechanisms, and finally PRS construction for risk prediction. This integrated approach leverages the strengths of each method while mitigating their individual limitations.
Table 3: Essential Research Reagents and Computational Tools
| Category | Item | Specification/Version | Function | Application Notes |
|---|---|---|---|---|
| Software Packages | PRS-CSx | Python implementation | Bayesian polygenic prediction | Requires LD reference panels |
| genMOSS | R package | Bayesian graphical models | MCMC for high-dimensional space | |
| ARTP2 | R package | Pathway enrichment analysis | Accepts summary statistics | |
| BGLR | R package | Bayesian regression models | Implements Bayesian Alphabet | |
| Reference Data | 1000 Genomes | Phase 3 | LD reference panels | Multi-ancestry foundation |
| GWAS Catalog | Current release | Prior knowledge base | Informed priors for Bayesian methods | |
| KEGG/Reactome | Current release | Pathway definitions | Biological context for ARTP2 | |
| Quality Control | PLINK | v1.9/v2.0 | Genotype processing | Data preprocessing and QC |
| R | v4.0+ | Statistical computing | Primary analysis environment |
The integration of PRS-CSx, Bayesian modeling, and Adaptive Rank Truncated Product methods represents a significant advancement in trans-ancestry endometriosis research. These core analytical frameworks address critical challenges in complex disease genetics, including population diversity, polygenic architecture, and biological interpretation. By providing detailed protocols and implementation guidelines, this article enables researchers to apply these sophisticated methods to advance our understanding of endometriosis genetics across diverse global populations.
Future methodological developments will likely focus on enhancing cross-population portability, integrating multi-omics data, and improving computational efficiency for large-scale biobank data. As these methods evolve, they will continue to transform endometriosis research, ultimately contributing to improved risk prediction, clinical stratification, and targeted therapeutic development for this complex gynecological disorder.
Multi-ancestry genome-wide association studies (GWAS) represent a transformative approach in genetic epidemiology, addressing historical biases toward European-ancestry populations that have limited the generalizability of genetic discoveries [32]. By integrating data from diverse ancestral backgrounds, researchers can leverage differences in linkage disequilibrium (LD) patterns, allele frequencies, and genetic architectures to enhance variant discovery, improve fine-mapping resolution, and develop more portable polygenic risk scores [32] [33]. This Application Note provides detailed methodologies for implementing multi-ancestry GWAS approaches, with specific application to endometriosis research, a complex gynecological condition affecting approximately 10% of reproductive-aged women worldwide [12] [6].
The strategic integration of diverse genetic data addresses crucial limitations of single-ancestry studies while unlocking new biological insights. For endometriosis, recent multi-ancestry efforts in approximately 1.4 million women have identified 80 genome-wide significant associations, 37 of which are novel, demonstrating the substantial discovery potential of diverse cohorts [12]. Furthermore, cross-ancestry fine-mapping has proven particularly valuable for narrowing candidate causal variants within associated loci, with studies reporting 19 of 113 independent signals pinpointed within 95% credible sets [33].
Two primary computational strategies dominate multi-ancestry GWAS implementations, each with distinct advantages and considerations for endometriosis research.
Pooled analysis combines individual-level genetic data from all ancestral backgrounds into a single unified dataset, typically incorporating principal components or mixed-effects models to account for population stratification [32] [34]. This approach maximizes statistical power through increased sample size and efficiently handles admixed individuals without requiring arbitrary ancestry categorizations [35].
Recent evaluations demonstrate that pooled analysis generally provides superior statistical power compared to meta-analysis approaches across various ancestry compositions and trait architectures, particularly when allele frequencies differ across populations [32] [34]. The method maintains well-controlled type I error rates in realistic scenarios with proper stratification control [35]. Implementation typically employs mixed-effect models (e.g., REGENIE) to account for population structure and relatedness, especially important in biobank-scale datasets where cryptic relatedness is common [32].
Meta-analysis conducts separate GWAS within defined ancestry groups and subsequently combines summary statistics using fixed-effect or random-effects models [32] [36]. This approach effectively captures fine-scale population structure within homogeneous groups and facilitates data sharing when individual-level data are restricted [34].
Advanced meta-analysis extensions like MR-MEGA leverage allele-frequency differences among contributing studies to enhance power and handle admixed individuals [32]. However, this method introduces additional parameters that can reduce power, particularly with complex admixture patterns [32]. Limitations include reduced effectiveness of population structure correction in smaller cohorts and potential exclusion of individuals who don't fit neatly into predefined ancestry categories [32] [36].
Table 1: Comparison of Multi-ancestry GWAS Methodological Approaches
| Feature | Pooled Analysis | Meta-Analysis | MR-MEGA |
|---|---|---|---|
| Data Structure | Individual-level data combined | Summary statistics combined | Summary statistics combined with ancestry parameters |
| Population Structure Control | Principal components, mixed models | Within-group corrections | Leverages allele frequency differences |
| Handling of Admixed Individuals | Direct inclusion | Challenging, often excluded | Specifically designed for admixture |
| Statistical Power | Generally higher [32] [34] | Moderate | Variable, reduced with complex admixture [32] |
| Implementation Complexity | Higher computational demands | Lower, facilitates distributed analysis | Moderate, requires careful parameterization |
| Data Sharing Considerations | Requires individual data access | Can use summary statistics | Can use summary statistics |
Empirical evaluations across multiple biobanks demonstrate the practical implications of method selection. In analyses of eight continuous and five binary traits from the UK Biobank (N â 324,000) and All of Us Research Program (N â 207,000), pooled analysis consistently exhibited better statistical power while effectively controlling for population stratification [34] [35]. Similarly, in the Hyperglycemia and Adverse Pregnancy Outcome Study, heterogeneous ancestry mega-analysis identified significantly more associations with maternal glucose measures compared to homogeneous ancestry meta-analysis, including biologically credible signals at the MTNR1B locus that were missed by the meta-analysis approach [36].
Graph 1: Meta-analysis workflow for multi-ancestry GWAS. The process involves ancestry stratification, group-specific imputation and association testing, followed by summary statistics harmonization and cross-ancestry integration.
Endometriosis exhibits substantial heritability, yet previous GWAS have explained only ~5% of disease variance [2]. Recent multi-ancestry efforts have dramatically expanded our understanding, with the largest study to date (N â 1.4 million women) identifying 80 genome-wide significant associations, including 37 novel loci and 5 inaugural adenomyosis variants [12]. Functional annotation revealed enrichment in pathways involved in immune regulation, tissue remodeling, and cell differentiation, providing mechanistic insights into disease pathogenesis [12].
Graph 2: Comprehensive analytical framework for multi-ancestry endometriosis research, integrating genetic discovery with functional annotation and therapeutic prioritization.
Multi-ancestry endometriosis studies have successfully identified potential therapeutic targets through Mendelian randomization and colocalization analysis. Recent research implicated RSPO3 and FLT1 as potential therapeutic candidates, with external validation confirming robust associations for RSPO3 [6]. Drug-repurposing analyses have highlighted interventions currently used for breast cancer and preterm birth prevention as promising candidates [12].
Table 2: Key Research Reagent Solutions for Multi-ancestry Endometriosis GWAS
| Reagent/Resource | Function | Specifications | Application in Endometriosis |
|---|---|---|---|
| TOPMed Freeze 8 | Cosmopolitan reference panel | Diverse ancestries, whole genome sequencing | Unified imputation for heterogeneous cohorts [36] [33] |
| Multi-Ethnic Genotyping Array (MEGA) | Genome-wide variant screening | ~2M markers optimized for diverse populations | Initial genotyping of multi-ancestry cohorts [36] |
| SOMAscan V4 | Plasma protein quantification | 4,907 protein targets | pQTL mapping for therapeutic target identification [6] |
| REGENIE | Mixed-model GWAS | Handles relatedness, population structure | Association testing in pooled analyses [32] |
| FUMA | Functional annotation | Integrates multiple genomic databases | Prioritization of endometriosis-associated loci [37] |
| PoPS | Gene prioritization | Polygenic Priority Score algorithm | Identification of endometriosis effector genes [33] |
Multi-ancestry GWAS integration represents a paradigm shift in endometriosis genetics, enhancing discovery power and biological insights while promoting health equity. The methodological framework outlined in this Application Note provides researchers with standardized protocols for implementing these approaches, from initial study design through functional interpretation. As diverse biobanks continue to expand, these strategies will be essential for translating genetic discoveries into clinically actionable insights for endometriosis diagnosis, treatment, and prevention.
The integration of multi-ancestry genome-wide association studies (GWAS) has become crucial for advancing our understanding of complex diseases like endometriosis. Endometriosis, affecting approximately 5-10% of reproductive-age women, is now recognized as a systemic inflammatory disease rather than merely a localized pelvic condition [38]. Its etiopathogenesis involves a complex interplay between genetic inheritance and environmental influences, with GWAS having identified numerous disease risk loci [38]. However, traditional single-ancestry genetic studies have limitations in generalizability and power, creating an urgent need for sophisticated trans-ancestry integration methods that can leverage diverse genetic datasets [39].
This protocol details three complementary analytical frameworksâSNP-centric, gene-centric, and pathway-centric approachesâfor integrating trans-ancestry genetic data in endometriosis research. Each method offers distinct advantages for aggregating association signals across different ancestral populations, including African, East Asian, and European cohorts [39]. By implementing these strategies, researchers can enhance detection efficiency, improve biological interpretation, and identify novel therapeutic targets for this complex gynecological disorder.
The trans-ancestry integration framework operates under the Trans-Ancestry Gene Consistency (TAGC) assumption, which posits that a specific subset of genes within a pathway is associated with endometriosis across various ancestry groups, though association strengths may differ due to genetic and environmental variations [39]. This assumption is biologically plausible since functional variants, particularly common ones, are often shared among diverse populations [39]. The integration strategies are categorized by the level at which genetic data is combined: SNP-level, gene-level, or pathway-level.
Table 1: Comparison of Trans-Ancestry Integration Approaches
| Approach | Integration Level | Key Methodology | Primary Advantage | Best Use Case |
|---|---|---|---|---|
| SNP-Centric | Individual SNPs | Consolidates SA-SNP summary statistics to generate TA-SNP statistics [39] | Maximizes fine-mapping resolution | Identifying specific causal variants |
| Gene-Centric | Gene-level | Aggregates SA-SNP data within genes to produce SA-gene statistics, then unifies across ancestries [39] | Balances resolution and biological interpretability | Candidate gene prioritization |
| Pathway-Centric | Pathway-level | Integrates p-values from pathway analyses across each SA-GWAS [39] | Captures polygenic effects across biological systems | Pathway identification and therapeutic targeting |
The SNP-centric approach begins with consolidating single-ancestry SNP-level (SA-SNP) summary data from multiple genome-wide association studies to generate trans-ancestry SNP-level (TA-SNP) summary statistics [39].
Step 1: Data Preparation and Harmonization
Step 2: Effect Size Modeling
Step 3: Trans-Ancestry SNP Statistic Calculation
Z_TA = (Σ w_i * Z_i) / â(Σ w_i²) where wi = 1/SEi [39]Step 4: Gene-Level Aggregation
For endometriosis research, incorporate genomic predictors beyond GWAS summary statistics, including:
Validate SNP-gene assignments using endometriosis-relevant tissues (uterus, endometrium) from GTEx v8 dataset [10].
The gene-centric approach first aggregates single-ancestry SNP data within genes, then unifies these gene-level statistics across ancestry groups [39].
Step 1: Single-Ancestry Gene-Level Analysis
Step 2: Gene-Level Statistics Integration
Step 3: Biological Validation and Prioritization
Gene-centric integration has identified specific endometriosis-risk genes including:
The pathway-centric approach conducts pathway analysis separately for each ancestry group, then integrates the results across populations [39].
Step 1: Single-Ancestry Pathway Analysis
Step 2: Pathway P-value Integration
Step 3: Endometriosis-Specific Pathway Enrichment
Table 2: Endometriosis-Relevant Pathways Identified Through Trans-Ancestry Integration
| Pathway Category | Specific Pathways | Biological Significance in Endometriosis | Therapeutic Implications |
|---|---|---|---|
| Inflammatory Processes | Neutrophil Degranulation [38] | Facilitates metastasis-like spread to distant organs | Potential for immunomodulator repurposing |
| Hormone Metabolism | Estrogen Receptor Signaling [38] | Drives lesion establishment and growth | ESR1-targeting agents in clinical trials |
| Cell Processes | PI3K/AKT/mTOR [38], Cell Adhesion/Migration [2] | Promotes lesion survival and invasion | AKT1 inhibitors, anti-adhesion therapies |
| Stress Response | Autophagy [2] | Supports cell survival in ectopic locations | Novel therapeutic target |
| Immune Function | Macrophage Biology [2] | Creates pro-inflammatory microenvironment | Immunomodulatory approaches |
Table 3: Key Research Reagent Solutions for Trans-Ancestry Endometriosis Studies
| Resource Category | Specific Resource | Function in Analysis | Access Information |
|---|---|---|---|
| GWAS Summary Data | UK Biobank (ukb-b-10903) [6] | Endometriosis case-control data | https://www.ukbiobank.ac.uk/ |
| Multi-Ancestry Data | FinnGen R10/R12 [10] [6] | Validation cohorts for trans-ancestry analysis | https://www.finngen.fi/en |
| QTL Databases | eQTLGen [10], GTEx v8 [10] | Expression quantitative trait loci data | https://www.eqtlgen.org/ |
| Pathway Databases | KEGG, MSigDB [38] | Curated biological pathways for enrichment analysis | https://www.genome.jp/kegg/ |
| Analysis Tools | SMR software [10], ARTP method [39] | Multi-omic integration and pathway analysis | https://cnsgenomics.com/software/smr/ |
| Prior Knowledge Bases | CellAge [10], STRING [38] | Cellular aging genes and protein interaction networks | https://genomics.senescence.info/cells/ |
| Methaniminium | Methaniminium, CAS:53518-13-1, MF:CH4N+, MW:30.049 g/mol | Chemical Reagent | Bench Chemicals |
| 1-Hexadecen-3-one | 1-Hexadecen-3-one|CAS 42459-63-2|C16H30O | High-purity 1-Hexadecen-3-one (C16H30O) for semiochemical and ecological research. This product is for Research Use Only (RUO). Not for human or veterinary use. | Bench Chemicals |
Sample Size Requirements
Quality Control Measures
Endometriosis-Specific Considerations
The integration of SNP-centric, gene-centric, and pathway-centric approaches provides a powerful framework for advancing trans-ancestry endometriosis research. By leveraging diverse genomic datasets and multi-omic integration strategies, researchers can overcome limitations of single-ancestry studies, enhance discovery power, and identify biologically relevant mechanisms driving endometriosis pathogenesis. The protocols outlined here provide a roadmap for implementing these approaches, with specific considerations for endometriosis applications. As multi-ancestry resources continue to expand, these methods will become increasingly essential for translating genetic discoveries into improved diagnostics and therapeutics for this complex gynecological disorder.
The integration of transcriptome-wide and proteome-wide association studies represents a transformative approach in complex disease research, enabling the identification of functionally relevant molecular mechanisms that transcend genomic associations alone. This integrated framework is particularly powerful when applied to endometriosis, a heritable inflammatory condition affecting 5-10% of reproductive-aged women worldwide, with an estimated heritability of 47-52% [19] [41]. While genome-wide association studies (GWAS) have successfully identified multiple risk loci for endometriosis, these predominantly lie in non-coding regions, suggesting regulatory functions that can only be fully elucidated through multi-omics integration [41]. This protocol details comprehensive methodologies for trans-ancestry meta-analysis coupled with transcriptomic and proteomic profiling to bridge the gap between genetic susceptibility and functional pathophysiology in endometriosis research.
Table 1: Key Findings from Multi-Omics Endometriosis Studies
| Study Type | Sample Size | Key Quantitative Findings | Significance |
|---|---|---|---|
| GWAS Meta-analysis [21] | 17,045 cases; 191,596 controls | 5 novel loci (FN1, CCDC170, ESR1, SYNE1, FSHB); 19 independent SNPs explaining â¤5.19% variance | P < 5 à 10-8; highlights genes in sex steroid hormone pathways |
| Proteomics [42] | 39 samples across cohorts | 73,218 tryptic peptides; 8,032 unique proteins quantified; 41 ubiquitinated fibrosis-related proteins identified | Proteins with FC >1.5, p < 0.05 considered significant |
| Ubiquitylomics [42] [43] | 5 normal; 6 EU/EC pairs | 1,647 ubiquitinated lysine sites (EC vs NC); 1,698 sites (EC vs EU); 8,407 Kub peptides total | Correlation coefficients: 0.32 (EC/NC) and 0.36 (EC/EU) for ubiquitinated fibrosis proteins |
| Transcriptomics [42] | 6 NC; 6 EU; 10 EC | 41 differentially expressed genes in menstrual stem cells; 16,383 characterized transcripts | FDR < 0.1; genes involved in proliferation, migration, steroid response |
| Multi-omics SMR [10] | 21,779 cases; 449,087 controls | 196 CpG sites in 78 genes; 18 eQTL-associated genes; 7 pQTL-associated proteins | PSMR < 0.05; PHEIDI > 0.05; PPH4 > 0.5 for colocalization |
Table 2: Experimentally Validated Molecular Targets in Endometriosis
| Target Category | Specific Molecules | Expression/Function in Endometriosis | Experimental Validation |
|---|---|---|---|
| Fibrosis-Related Proteins | TGFBR1, α-SMA, FAP, FN1, Collagen1 | Elevated in ectopic lesions [42] | Western blot across independent samples |
| E3 Ubiquitin Ligase | TRIM33 | mRNA and protein levels reduced in endometriotic tissues [42] [43] | siRNA knockdown in hESCs promoted TGFBR1/p-SMAD2/α-SMA/FN1 |
| Extracellular Matrix Components | COL1A1, COL6A2, LAMC3, NID2 | Dysregulated in endometriosis MenSCs [44] | Proteomic analysis (UPLC-MS/MS) with p < 0.05 |
| Transcription Factors | ATF3, ID1, ID3, FOSB, SNAI1, NR4A1 | Protein-protein interaction enrichment (p < 1.0 Ã 10-16) [44] | RNA-seq of menstrual mesenchymal stem cells |
Principle: Large-scale meta-analysis of genome-wide association studies across diverse populations enhances power to detect risk loci and enables fine-mapping of causal variants [21] [19].
Sample Requirements:
Quality Control Steps:
Imputation:
Association Analysis:
Meta-Analysis:
Downstream Analysis:
Figure 1: Trans-ancestry GWAS meta-analysis workflow detailing cohort preparation through to risk loci identification
Principle: Parallel RNA sequencing and proteomic analysis of matched tissues identifies concordant molecular pathways and reveals post-transcriptional regulatory mechanisms in endometriosis pathogenesis [42] [44].
Sample Collection and Preparation:
RNA Sequencing:
Proteomic Analysis (DIA-PASEF):
Multi-Omics Integration:
Principle: Comprehensive identification of ubiquitination sites reveals post-translational regulatory mechanisms in endometriosis fibrosis [42] [43].
Sample Preparation:
Ubiquitinated Peptide Enrichment:
LC-MS/MS Analysis:
Data Processing:
Functional Validation:
Figure 2: Multi-omics integration framework connecting genetic variants to molecular and clinical phenotypes
Table 3: Essential Research Reagents for Multi-Omics Endometriosis Studies
| Reagent Category | Specific Products | Application | Key Features |
|---|---|---|---|
| Nucleic Acid Extraction | TRIzol Reagent (Magen Biotech) [42] | RNA isolation for transcriptomics | Maintains RNA integrity; compatible with multiple sample types |
| Library Preparation | ABclonal mRNA-seq Lib Prep Kit [42] | RNA-seq library construction | Poly-A selection; fragmentation optimization; dual-index barcoding |
| Proteomics Digestion | Sequencing-grade trypsin [42] | Protein digestion for MS | High specificity; low autolysis; compatible with ubiquitination studies |
| Ubiquitin Enrichment | Anti-K-ε-GG antibody beads [42] | Ubiquitylomics profiling | High-affinity antibody; specific ubiquitinated peptide enrichment |
| Chromatography | UHPLC systems (EASY-nLC) [42] [44] | Peptide separation | Nanoflow capabilities; high reproducibility; acetonitrile gradients |
| Mass Spectrometry | TimSTOF Pro (Bruker); Q-Exactive HF-X (Thermo) [42] | Proteomic/ubiquitylomic analysis | High resolution; PASEF capability; high sensitivity |
| Cell Culture | Primary human endometrial stromal cells (hESCs) [42] [43] | Functional validation | Primary cell model; relevant pathophysiology |
| Gene Silencing | TRIM33 siRNA [42] [43] | E3 ligase functional studies | Target-specific; high knockdown efficiency |
| Validation Antibodies | Anti-TGFBR1, anti-α-SMA, anti-FN1 [42] | Western blot confirmation | Target-specific; validated for endometriosis tissues |
| Neodymium;ZINC | Neodymium;ZINC Research Compound|NdZn | Neodymium;ZINC (NdZn5) for research applications in materials science, agriculture, and catalysis. This product is For Research Use Only (RUO), not for human or veterinary use. | Bench Chemicals |
Multi-Omics Integration Pipeline:
Colocalization Analysis:
Pathway and Network Analysis:
Quality Control Metrics:
The integrated application of transcriptome-wide and proteome-wide association studies within trans-ancestry meta-analysis frameworks provides unprecedented resolution for elucidating endometriosis pathogenesis. The experimental protocols detailed herein enable researchers to bridge the gap between genetic susceptibility and functional pathophysiology, with particular emphasis on post-translational regulatory mechanisms such as ubiquitination that drive critical disease processes like fibrosis. The standardized methodologies, analytical frameworks, and reagent solutions presented offer a comprehensive toolkit for advancing our understanding of endometriosis and identifying novel therapeutic targets for this complex gynecological disorder.
The application of trans-ancestry meta-analysis methods in genome-wide association studies (GWAS) for endometriosis represents a transformative approach for identifying novel therapeutic targets and enabling drug repurposing opportunities. Endometriosis, a common gynecological disorder affecting approximately 10% of reproductive-age women globally, demonstrates significant genetic underpinnings with a heritability estimated at around 50% [45] [2]. Despite this strong genetic component, traditional GWAS approaches have explained only a limited fraction of disease variance, highlighting the need for more sophisticated analytical frameworks that can integrate diverse ancestral datasets to improve statistical power and resolution [2] [12].
Drug repurposingâidentifying new therapeutic uses for existing medicationsâhas emerged as an economically efficient strategy that leverages established safety profiles to accelerate treatment development. The average cost to market a repurposed drug is approximately $300 million, substantially less than the $2â3 billion typically required for novel drug development [46]. Genetic evidence significantly enhances this process, with drug mechanisms supported by human genetic evidence demonstrating a 2.6 times greater probability of clinical success compared to those without such support [47]. This review presents integrated protocols and application notes for conducting drug repurposing analyses within the context of trans-ancestry endometriosis research, providing researchers with methodological frameworks for translating genetic discoveries into therapeutic hypotheses.
The foundation of effective drug repurposing analyses begins with robust trans-ancestry genetic study design. Recent advancements have demonstrated the power of large-scale, diverse cohorts in endometriosis genetics. A 2025 multi-ancestry GWAS of approximately 1.4 million women, including 105,869 endometriosis cases, identified 80 genome-wide significant associations, 37 of which were novel [12]. This study established the feasibility of expanding endometriosis locus discovery across ancestries while enabling the dissection of symptom-specific genetic effects.
Table 1: Summary of Recent Endometriosis Genetic Studies Utilizing Multi-Ancestry Approaches
| Study | Sample Size | Cases | Key Findings | Ancestries Represented |
|---|---|---|---|---|
| Multi-ancestry GWAS (2025) [12] | ~1.4 million women | 105,869 | 80 significant loci (37 novel), 5 first adenomyosis variants | Multiple, unspecified |
| Taiwanese-Han GWAS (2024) [45] | 30,734 | 2,794 | 5 significant susceptibility loci (2 novel) | Taiwanese-Han |
| Combinatorial Analysis (2025) [2] | UK Biobank + All of Us | Unspecified | 1,709 disease signatures comprising 2,957 unique SNPs | White European, multi-ancestry validation |
The integration of datasets across diverse populations requires careful consideration of population stratification, imputation quality, and ancestral representation. The Taiwanese-Han GWAS exemplifies the value of population-specific analyses, identifying novel susceptibility loci while replicating known associations from European and Japanese cohorts [45]. Such studies highlight both shared genetic architecture across populations and population-specific risk factors that may inform targeted therapeutic development.
Materials:
Procedure:
This workflow enables improved fine-mapping resolution by leveraging differential linkage disequilibrium patterns across populations, potentially narrowing candidate causal variants from hundreds to single digits at associated loci [12].
Following the identification of association signals through trans-ancestry meta-analysis, the next critical step involves prioritizing genes with the greatest potential as therapeutic targets. Multi-omic integration approaches have demonstrated particular utility in this process. A 2025 study integrating transcriptomic, epigenetic, and proteomic data revealed that genetic variation influences endometriosis risk through regulation across multiple tissues, converging on pathways involved in immune regulation, tissue remodeling, and cell differentiation [12].
Table 2: Experimentally Validated Endometriosis Drug Repurposing Candidates
| Target | Drug Candidate | Evidence Level | Proposed Mechanism | Source |
|---|---|---|---|---|
| RSPO3 | Not specified | MR + experimental validation | Causal role in endometriosis pathogenesis | [6] |
| FLT1 | Not specified | MR + experimental validation | Potential involvement in vascular function | [6] |
| Multiple novel genes | Multiple possibilities | Combinatorial analytics | Pathways including autophagy and macrophage biology | [2] |
Mendelian randomization (MR) analysis has emerged as a powerful method for establishing causal relationships between putative targets and endometriosis risk. A recent MR study investigating blood metabolites and plasma proteins identified RSPO3 and FLT1 as potentially causally associated with endometriosis [6]. Subsequent experimental validation through ELISA, RT-qPCR, and Western blotting confirmed elevated RSPO3 levels in both plasma and lesion tissues of endometriosis patients compared to controls [6].
Materials:
Procedure:
This MR framework establishes genetic support for target-disease relationships, which corresponds to a 2.6-fold greater probability of clinical success compared to non-genetically supported targets [47].
Traditional GWAS approaches have explained only approximately 5% of endometriosis disease variance, highlighting the limitations of single-variant analysis frameworks [2]. Combinatorial analytics represents a paradigm shift by examining how multiple genetic variants interact to influence disease risk. A 2025 study applying combinatorial analytics to endometriosis identified 1,709 disease signatures comprising 2,957 unique SNPs in combinations of 2-5 SNPs [2]. These signatures demonstrated high reproducibility (58-88%) in multi-ancestry validation cohorts, with reproducibility rates reaching 80-88% for higher frequency signatures (>9% frequency) [2].
Pathway enrichment analysis of these combinatorial signatures revealed involvement in biologically relevant processes including cell adhesion, proliferation and migration, cytoskeleton remodeling, angiogenesis, fibrosis, and neuropathic pain [2]. Notably, this approach identified 75 novel gene associations not previously linked to endometriosis through GWAS, highlighting its potential for uncovering new biology and therapeutic opportunities.
Materials:
Procedure:
The high reproducibility rates of combinatorial signatures across diverse ancestries (66-76% in non-white European sub-cohorts) underscores their utility in trans-ancestry drug repurposing analyses [2].
Table 3: Essential Research Reagents for Endometriosis Drug Repurposing Studies
| Reagent/Category | Specific Examples | Function/Application | Evidence Source |
|---|---|---|---|
| Protein Detection | Human R-Spondin3 ELISA Kit | Quantitative measurement of RSPO3 protein levels in patient plasma | [6] |
| Gene Expression Analysis | RT-qPCR reagents | Validation of gene expression differences in patient tissues | [6] |
| Genetic Datasets | UK Biobank, FinnGen, Taiwan Biobank | Source of GWAS summary statistics and individual-level genetic data | [45] [12] [6] |
| Protein QTL Resources | deCODE GWAS, SOMAscan data | Genetic instruments for Mendelian randomization analyses | [46] [6] |
| Pathway Analysis Tools | GO, KEGG, Reactome databases | Functional annotation of candidate genes and enrichment testing | [2] |
| Computational Platforms | PrecisionLife combinatorial analytics | Identification of multi-SNP disease signatures | [2] |
The integration of trans-ancestry meta-analysis methods with sophisticated drug repurposing frameworks presents unprecedented opportunities for accelerating therapeutic development in endometriosis. The protocols and application notes outlined herein provide researchers with comprehensive methodologies for translating genetic discoveries across diverse populations into clinically actionable therapeutic hypotheses. The remarkable reproducibility of combinatorial disease signatures across ancestries [2], coupled with the robust clinical success advantage for genetically supported drug targets [47], underscores the transformative potential of these approaches.
Future directions in this field will likely focus on expanding diverse ancestral representation in genetic studies, deepening multi-omic integration, and developing more sophisticated in silico models of drug-target interactions. As these methodologies mature, they will increasingly enable precision medicine approaches in endometriosis treatment, potentially targeting specific molecular subtypes across different ancestral backgrounds. The continuing growth of genetic datasets and analytical innovations promises to further accelerate the identification of repurposing opportunities, ultimately reducing the diagnostic and therapeutic delays that have long plagued endometriosis patients.
Endometriosis is a common, complex gynecological disorder influenced by multiple genetic and environmental factors, with an estimated heritability of approximately 51% based on twin studies [19]. Genome-wide association studies (GWAS) have successfully identified numerous genetic loci associated with endometriosis risk, revealing a polygenic architecture characterized by significant heterogeneity across ancestral populations [19] [21]. This application note examines the genetic architecture heterogeneity in endometriosis, focusing on effect size variations and linkage disequilibrium (LD) patterns across populations, and provides detailed protocols for trans-ancestry meta-analysis methods to enhance discovery and validation of risk loci.
GWAS meta-analyses have identified multiple genomic regions associated with endometriosis risk. The table below summarizes key loci and their heterogeneous effects across populations:
Table 1: Effect Size Variations of Endometriosis Risk Loci Across Populations
| Locus/Nearest Gene | Chromosome | Lead SNP | Effect Size (OR) European | Effect Size (OR) Japanese | P-Value | Key Biological Pathway |
|---|---|---|---|---|---|---|
| WNT4 | 1p36.12 | rs7521902 | 1.16 | 1.20 | 4.6Ã10â»â¸ | Development, steroidogenesis |
| GREB1 | 2p25.1 | rs13394619 | 1.14 | 1.08 | 6.1Ã10â»â¸ | Cell growth, estrogen regulation |
| Intergenic | 2p14 | rs4141819 | 1.12 | 1.11 | 8.5Ã10â»â¸ | Unknown |
| ID4 | 6p22.3 | rs7739264 | 1.11 | 1.10 | 3.6Ã10â»Â¹â° | Development, differentiation |
| Intergenic | 7p15.2 | rs12700667 | 1.22 | 1.22 | 9.3Ã10â»Â¹â° | Unknown |
| CDKN2B-AS1 | 9p21.3 | rs1537377 | 1.10 | 1.09 | 2.4Ã10â»â¹ | Cell cycle regulation |
| VEZT | 12q22 | rs10859871 | 1.13 | 1.14 | 5.1Ã10â»Â¹Â³ | Cell adhesion |
| FN1 | 2q35 | rs1250241 | 1.23* | - | 2.99Ã10â»â¹ | Extracellular matrix |
| ESR1 | 6q25.1 | rs1971256 | 1.09 | - | 3.74Ã10â»â¸ | Estrogen receptor |
| FSHB | 11p14.1 | rs74485684 | 1.11 | - | 2.00Ã10â»â¸ | Hormone regulation |
*Effect sizes marked with * are for Stage III/IV (Grade B) endometriosis only [19] [21] [48].
Trans-ancestry analyses reveal distinct patterns of heterogeneity. The CDKN2B-AS1 locus (rs10965235) exemplifies population-specific effects, demonstrating a substantial effect (OR=1.44) in Japanese populations but being monomorphic in European populations [48]. Conversely, the WNT4 locus shows consistent effects across European (OR=1.16) and Japanese (OR=1.20) ancestries [48]. A notable finding is that most loci exhibit stronger effect sizes in Stage III/IV endometriosis, suggesting they primarily influence the development of moderate to severe disease [19].
Table 2: Heterogeneity Metrics for Key Endometriosis Loci
| Locus | Cochran's Q P-value | I² Statistic | Effect Size Difference EUR vs. JPN | Consistent Direction Across Populations |
|---|---|---|---|---|
| 2p14 (rs4141819) | <0.005 | 78.3% | 0.01 | Yes |
| 2p25.1 (rs13394619) | 0.12 | 45.2% | 0.06 | Yes |
| 7p15.2 (rs12700667) | 0.87 | 0% | 0.00 | Yes |
| 12q22 (rs10859871) | 0.23 | 29.7% | -0.01 | Yes |
Table 3: Essential Research Reagents and Computational Tools
| Item | Specification | Function/Application |
|---|---|---|
| Genotyping Array | Illumina Global Screening Array, Affymetrix Axiom Biobank Array | Genome-wide SNP genotyping |
| Imputation Reference Panel | 1000 Genomes Project Phase 3, TOPMed | Genotype imputation to increase variant coverage |
| Quality Control Software | PLINK 2.0, QCTOOL, SNPTEST | Data filtering, quality control, and format conversion |
| Ancestry Determination Software | ADMIXTURE, EIGENSOFT | Population structure analysis and ancestry assignment |
| Summary Statistics | GWAS Catalog, EBI Biobank | Access to published endometriosis GWAS data |
Cohort Selection and Ancestry Stratification
Genotype Quality Control
Genotype Imputation
Figure 1: Trans-ancestry Meta-analysis Workflow
Effect Size Harmonization
Fixed-Effects Meta-Analysis
Heterogeneity Quantification
Genetic Correlation Analysis
Variant Prioritization
Colocalization Analysis
Polygenic Risk Score Assessment
The 7p15.2 locus (rs12700667) provides an exemplary case of consistent genetic effects across populations. Initial discovery in European populations (OR=1.20, P=1.4Ã10â»â¹) [48] showed successful replication in Japanese populations (OR=1.22, P=3.6Ã10â»Â³) [48]. The trans-ancestry meta-analysis bolstered the significance (P=9.3Ã10â»Â¹â°) with no evidence of heterogeneity (I²=0%) [19] [48].
In contrast, the 2p14 locus (rs4141819) exhibited significant heterogeneity (P<0.005) [19], suggesting potential population-specific causal variants or interactions with environmental factors. This heterogeneity necessitates careful interpretation in cross-population genetic risk prediction.
Mendelian randomization (MR) analyses have revealed causal relationships between serum lipid levels and endometriosis risk, particularly for triglycerides (TG) [22]. Drug-target MR has identified potential therapeutic targets including LPL, PPARA, ANGPTL3, and APOC3 [22].
Figure 2: Mendelian Randomization Reveals Causal Pathways
Integration of endometriosis gene expression data has revealed dysregulated pathways in ectopic lesions, including:
Phylogenetic analysis of gene expression patterns demonstrates that endometriosis lesions represent clonal outgrowths with accumulated genetic and epigenetic alterations [50], highlighting the importance of considering lesion heterogeneity in molecular studies.
Addressing genetic architecture heterogeneity is crucial for advancing endometriosis research. The protocols outlined herein enable robust trans-ancestry meta-analysis that accounts for effect size variations and LD patterns across populations. Key considerations include:
These approaches facilitate the development of more accurate polygenic risk scores across diverse populations and identify potential therapeutic targets for this complex gynecological disorder.
Trans-ancestry genome-wide association studies (GWAS) meta-analysis has emerged as a powerful approach for enhancing the discovery of genetic loci and improving the fine-mapping of causal variants for complex diseases. Within endometriosis research, this method is particularly valuable given the condition's high heritability (estimated at ~52%) and its global prevalence affecting 5-10% of reproductive-aged women [19]. However, the integration of datasets from diverse ancestral backgrounds introduces significant methodological challenges, primarily concerning population stratification and ancestry-specific confounders.
Population stratification occurs when differences in allele frequency between cases and controls arise from systematic ancestry differences rather than disease association. In trans-ancestry meta-analyses, this confounding can be substantially more pronounced than in single-ancestry studies due to the greater genetic diversity across populations. Failure to adequately account for these effects can produce spurious associations and reduce the portability of genetic risk scores across populations. This application note provides detailed protocols and analytical frameworks for managing these critical challenges specifically within the context of endometriosis GWAS research.
Principal Component Analysis remains a foundational approach for detecting and correcting population stratification. The method works by identifying the major axes of genetic variation in the dataset, which typically correspond to ancestral backgrounds.
Protocol: PCA Implementation for Trans-ancestry Endometriosis GWAS
In practice, studies such as the trans-ancestry meta-analysis of endometriosis that identified the WNT4 and GREB1 loci have successfully employed PCA to distinguish European and East Asian ancestry groups [48]. The variance explained by each PC should be carefully evaluated, with typically 5-10 PCs retained as covariates.
Genomic control and LD score regression provide complementary approaches to quantify and correct for residual population stratification.
Protocol: Genomic Inflation Assessment
For the endometriosis trans-ancestry meta-analysis by Painter et al., the genomic inflation factor was carefully monitored and reported, ensuring that the identified associations at loci such as 7p15.2 were not driven by stratification [19].
Fixed-effects and random-effects models present different advantages for trans-ancestry meta-analysis, with the choice dependent on between-population heterogeneity.
Protocol: Trans-ancestry Meta-analysis Implementation
Notably, the endometriosis meta-analysis by Rahmioglu et al. demonstrated remarkable consistency across populations for seven out of nine reported loci, supporting the use of fixed-effects models for these variants [19].
Table 1: Statistical Methods for Managing Population Stratification
| Method | Application | Advantages | Limitations |
|---|---|---|---|
| Principal Component Analysis | Correcting ancestry differences in combined datasets | Directly models continuous ancestry variation | May not capture fine-scale population structure |
| Genomic Control | Genome-wide correction of test statistics | Simple implementation | Overcorrection can reduce true positive signals |
| LD Score Regression | Quantifying inflation from stratification vs. polygenicity | Distinguishes biological signals from bias | Requires LD reference panels for each ancestry |
| Random-Effects Meta-analysis | Combining effects across heterogeneous populations | Conservative when heterogeneity is present | Reduced power compared to fixed-effects |
Robust ancestry determination forms the foundation of effective stratification control in trans-ancestry studies.
Protocol: Standardized Ancestry Reporting
The successful trans-ancestry endometriosis study by Painter et al. included 9,039 cases and 27,343 controls of European ancestry and 2,467 cases and 5,335 controls of Japanese ancestry, demonstrating the scale needed for well-powered detection [19].
Rigorous quality control must be applied both within and across ancestral groups to prevent technical artifacts from masquerading as biological signals.
Protocol: Trans-ancestry QC Pipeline
In the endometriosis GWAS by Albertsen et al., samples were restricted to those with â¥95% European ancestry based on ADMIXTURE analysis, reducing stratification concerns within the European cohort [51].
Table 2: Quality Control Thresholds for Trans-ancestry Endometriosis GWAS
| QC Metric | Threshold | Rationale | Tool Implementation |
|---|---|---|---|
| Sample Call Rate | >98% | Excludes poor-quality DNA samples | PLINK, QCtools |
| Variant Call Rate | >95% | Removes poorly genotyped markers | PLINK, VCFtools |
| Hardy-Weinberg Equilibrium | P > 1Ã10â»â¶ | Filters genotyping errors | PLINK, SNPTEST |
| Minor Allele Frequency | >1% | Ensures adequate power for association | PLINK, GENESIS |
| Heterozygosity | ±3SD from mean | Identifies sample contamination | PLINK, King |
| Relatedness | Ï < 0.2 | Prevents inflation from cryptic relatedness | King, PLINK |
The 2023 trans-ancestry meta-analysis of endometriosis provides an illustrative example of successfully implemented stratification controls [48]. This study integrated data from European and Japanese populations, specifically examining consistency and heterogeneity of genetic effects.
The analysis employed a multi-tiered approach to address population stratification:
The research identified significant heterogeneity at two independent inter-genic loci on chromosome 2 (rs4141819 and rs6734792), highlighting the importance of evaluating ancestry-specific effects rather than assuming uniform genetic architecture [19].
The meta-analysis confirmed six genome-wide significant loci with consistent effects across ancestries:
These findings demonstrated that despite ancestry differences, substantial sharing of genetic risk factors exists for endometriosis, providing a rationale for trans-ancestry approaches.
Table 3: Key Research Reagents for Trans-ancestry Endometriosis GWAS
| Reagent/Resource | Function | Example Implementation |
|---|---|---|
| Illumina Global Screening Array | Genotyping platform for diverse populations | Designed with content optimized for multi-ethnic studies, includes ancestry-informative markers |
| 1000 Genomes Project Reference | Population genetic reference panel | Provides allele frequency data across 26 populations for ancestry determination and imputation |
| TOPMed Imputation Reference | High-quality imputation panel | Improves variant discovery in diverse populations, enhances fine-mapping resolution |
| PLINK 2.0 | Whole-genome association analysis | Performs QC, PCA, and basic association testing with efficient handling of large datasets |
| METAL | Meta-analysis software | Combines GWAS results across studies with heterogeneity testing and multiple weighting schemes |
| LDAK | Heritability and stratification analysis | Estimates SNP heritability and performs LD-adjusted kinship analysis |
| GENESIS | Genetic association testing | Accounts for population structure and relatedness in diverse cohorts using mixed models |
| GTEx Database | Functional validation resource | Provides expression quantitative trait loci (eQTL) data for tissue-specific functional annotation |
Recent methodological advances offer promising approaches for further improving trans-ancestry genetic studies of endometriosis:
Multi-trait Analysis of GWAS (MTAG) MTAG enables efficient cross-population analysis by incorporating genetic correlations between ancestries, potentially increasing power for detecting endometriosis risk loci with heterogeneous effects.
Genetic Risk Prediction Methods Methods like PRS-CSx incorporate trans-ancestry information to improve polygenic risk prediction across diverse populations, addressing the current limitation where most PRS display reduced portability across ancestry groups.
The integration of functional genomics data represents a critical frontier for understanding ancestry-specific effects in endometriosis:
Expression Quantitative Trait Loci (eQTL) Mapping Studies such as the Taiwanese endometriosis GWAS identified eQTL effects, with rs13126673 showing association with INTU expression in endometriotic tissues (P = 0.034) [52]. Such findings highlight the importance of context-specific functional data.
Colocalization Analysis Bayesian colocalization methods can determine whether genetic associations with endometriosis and molecular traits (e.g., gene expression, DNA methylation) share causal variants, helping prioritize candidate genes across ancestries.
Effective management of population stratification and ancestry-specific confounders is essential for robust trans-ancestry endometriosis research. The protocols and methodologies outlined in this application note provide a comprehensive framework for addressing these challenges, from study design and quality control to advanced statistical analysis. As endometriosis genetics continues to advance toward more diverse and inclusive sampling, these approaches will be increasingly critical for ensuring that genetic discoveries translate across ancestral backgrounds and benefit all populations equally. The remarkable consistency observed across ancestries for most endometriosis risk loci provides strong justification for continued trans-ancestry efforts, which promise to further elucidate the genetic architecture of this complex gynecological condition.
Genomic research has revolutionized our understanding of complex diseases like endometriosis, yet significant disparities persist due to the underrepresentation of non-European populations in major biobanks and genome-wide association studies (GWAS). This ancestral bias creates substantial gaps in the equity and effectiveness of precision medicine approaches, particularly for conditions such as endometriosis that affect a global population. The current genomic databases, including The Cancer Genome Atlas (TCGA) and the GWAS Catalog, demonstrate a dramatic over-representation of individuals with European ancestry, with TCGA cancers having a median of 83% European ancestry individuals and the GWAS Catalog being approximately 95% European [53]. This imbalance severely limits the portability of genetic risk scores and therapeutic targets across diverse populations and restricts the fundamental understanding of disease biology that could be gained from analyzing ancestrally diverse genomic data.
The statistical consequences of this underrepresentation are profound. Model efficacy in genetic studies has been demonstrated to correlate directly with population sample size, meaning populations with little or no representation in training data experience larger disparities in disease model performance and garner minimal benefit from benchmark disease models [53]. Furthermore, European ancestry-based scores for genetic intolerance metrics are approaching saturation, meaning that simply adding more European-ancestry samples provides diminishing returns for variant discovery [54]. In contrast, increasing ancestral representation, rather than sample size alone, has been shown to critically drive the performance of key genomic metrics, with scores trained on African and Admixed American ancestral groups demonstrating higher resolution in detecting haploinsufficient and neurodevelopmental disease risk genes compared to scores trained on European ancestry groups [54]. For endometriosis research specifically, this ancestral bias presents a critical methodological challenge that requires specialized approaches to ensure equitable and statistically powerful research outcomes across all populations.
Table 1: Ancestral Representation in Major Genomic Databases
| Database/Resource | European Ancestry | African Ancestry | East Asian Ancestry | South Asian Ancestry | Admixed American | Citation |
|---|---|---|---|---|---|---|
| GWAS Catalog | ~95% | Not specified | Not specified | Not specified | Not specified | [53] |
| TCGA (median across cancers) | 83% (range 49-100%) | Not specified | Not specified | Not specified | Not specified | [53] |
| gnomAD v2 (exomes) | 56,885 (NFE) + 10,824 (Finnish) | 8,128 | 9,197 | 15,308 | 17,296 (Latino) | [54] |
| UK Biobank (exomes) | 437,812 (95.06%) | 8,701 (1.89%) | 2,150 (0.47%) | 9,217 (2.00%) | Not specified | [54] |
The representation disparities shown in Table 1 have direct consequences for statistical power. African ancestry cohorts exhibit approximately 1.8-fold enrichment of common missense variants compared to non-Finnish European cohorts, highlighting the substantial genetic diversity being missed in current studies [54]. This diversity is crucial for comprehensive gene discovery, as demonstrated by the fact that missense tolerance ratio (MTR) metrics trained on just 43,000 multi-ancestry exomes demonstrated greater predictive power than when trained on a nearly 10-fold larger dataset of 440,000 non-Finnish European exomes [54].
Table 2: Performance Comparison of Genetic Intolerance Metrics by Ancestry
| Ancestry Group | Sample Size (gnomAD) | AUC for NDD Genes (RVIS) | AUC for Haploinsufficient Genes | Fold Enrichment of Common Missense Variants | Citation |
|---|---|---|---|---|---|
| African (AFR) | 8,128 | Highest (0.71-0.85) | Moderate | 1.8x | [54] |
| Admixed American (AMR) | 17,296 | High | Moderate | Not specified | [54] |
| South Asian (SAS) | 15,308 | High | Moderate | Not specified | [54] |
| Non-Finnish European (NFE) | 56,885 | Lower than AFR | Moderate | Reference | [54] |
| Finnish (FIN) | 10,824 | Lower than AFR | Moderate | Lowest | [54] |
The data in Table 2 demonstrates that diverse ancestral representation significantly enhances the resolution of genic intolerance metrics. For instance, Residual Variance Intolerance Score (RVIS) metrics derived from African ancestry cohorts consistently achieved the highest area under the ROC curve (AUC) for detecting neurodevelopmental disorder (NDD) genes compared to European-based scores across multiple validation sets [54]. This pattern holds true despite the considerably smaller sample sizes for non-European groups, highlighting that diversity, rather than simply sample size, drives discovery power.
Purpose: To identify endometriosis risk loci with improved portability across diverse ancestral groups through trans-ethnic meta-analysis approaches.
Materials:
Procedure:
Ancestry-Specific Quality Control: Apply stringent quality control metrics separately for each ancestral group, including:
Trans-ethnic Fixed-Effects Meta-analysis: Perform fixed-effects meta-analysis using inverse-variance weighting to combine effects across ancestries:
Heterogeneity Assessment: Evaluate heterogeneity in effect sizes across ancestries using Cochran's Q statistic and I² values. Variants showing significant heterogeneity (p < 0.005) require careful interpretation in context of potential ancestry-specific effects [19].
Conditional Analysis for Secondary Signals: Identify independent association signals through stepwise conditional analysis within associated loci, as demonstrated in the identification of 19 independent SNPs for endometriosis [21].
Validation in Admixed Cohorts: Validate identified loci in admixed populations such as the All of Us cohort, which includes multi-ancestry participants [2].
Diagram Title: Trans-ethnic GWAS Meta-analysis Workflow
Purpose: To identify reproducible multi-SNP disease signatures across diverse ancestries using combinatorial analytics approaches.
Materials:
Procedure:
Combinatorial Association Testing: Implement combinatorial analytics to identify multi-SNP signatures associated with endometriosis risk:
Cross-ancestry Replication Testing: Test signatures identified in one ancestral group for replication in other ancestries:
Pathway Enrichment Analysis: Perform functional annotation of genes mapped from reproducing signatures using:
Novel Gene Prioritization: Prioritize novel genes identified through combinatorial approaches that do not overlap with previous GWAS findings, such as the 75 novel genes recently identified for endometriosis [2].
Purpose: To characterize the functional impact of endometriosis-associated variants through multi-tissue expression quantitative trait loci (eQTL) analysis.
Materials:
Procedure:
Tissue-specific eQTL Analysis: Cross-reference variants with eQTL data from six physiologically relevant tissues:
Significance Thresholding: Retain only significant eQTLs with false discovery rate (FDR) adjusted p-value < 0.05.
Effect Size Quantification: Extract slope values for significant eQTLs, representing the direction and magnitude of regulatory effects.
Functional Prioritization: Prioritize genes based on:
Hallmark Pathway Mapping: Map regulated genes to established biological pathways using:
Diagram Title: Multi-tissue eQTL Analysis Workflow
Table 3: Essential Research Reagents and Computational Tools
| Tool/Resource | Type | Primary Function | Application in Endometriosis Research | Citation |
|---|---|---|---|---|
| PhyloFrame | Computational Method | Equitable machine learning for genomic medicine | Corrects ancestral bias in disease signatures; improves predictions across ancestries for breast, thyroid, and uterine cancers | [53] |
| TEMR (Trans-ethnic MR) | Statistical Method | Mendelian randomization for underrepresented populations | Improves statistical power for causal inference in non-European populations using trans-ethnic genetic correlations | [55] |
| PrecisionLife Combinatorial Analytics | Analytical Platform | Identifies multi-SNP disease signatures | Discovered 1,709 endometriosis disease signatures with high cross-ancestry reproducibility (58-88%) | [2] |
| GTEx Database v8 | Data Resource | Multi-tissue gene expression reference | Enables eQTL mapping of endometriosis variants across uterus, ovary, colon, and blood tissues | [5] |
| PASS Software | Statistical Tool | Sample size and power analysis | Calculates required sample sizes for achieving sufficient statistical power (â¥0.90) in genetic studies | [56] |
| MONITOR Software | Statistical Tool | Power analysis for monitoring programs | Estimates statistical power for trend detection; adaptable to genetic study design | [57] |
| RVIS (Residual Variance Intolerance Score) | Genomic Metric | Gene-level intolerance to variation | Prioritizes candidate genes; African-ancestry versions show improved performance | [54] |
| MTR (Missense Tolerance Ratio) | Genomic Metric | Sub-genic intolerance to missense variation | Identifies protein domains intolerant to variation; benefits from diverse training data | [54] |
The integration of trans-ancestry genetic findings with functional genomics has revealed several key biological pathways in endometriosis pathogenesis that demonstrate consistency across diverse populations:
Novel endometriosis risk loci identified through trans-ethnic approaches implicate genes involved in sex steroid hormone pathways, including ESR1 (estrogen receptor 1), FSHB (follicle-stimulating hormone subunit beta), and CCDC170 (coiled-coil domain containing 170) [21]. These findings highlight the conserved role of hormonal regulation in endometriosis across ancestries. The ESR1 locus in particular contains multiple independent association signals identified through conditional analysis in trans-ethnic datasets [21].
Genes identified through combinatorial analytics approaches show strong enrichment in pathways involved in cell adhesion, proliferation, migration, and cytoskeleton remodeling [2]. These processes are fundamental to the establishment and survival of ectopic endometrial lesions. Multi-tissue eQTL analysis further demonstrates that endometriosis risk variants regulate key genes in these pathways, including FN1 (fibronectin 1) and CLDN23 (claudin 23), with effect sizes showing consistency across diverse populations [5].
Immune-related pathways predominate in the regulatory profiles of eQTL-associated genes in both peripheral blood and gastrointestinal tissues [5]. Key regulators such as MICB (MHC class I polypeptide-related sequence B) demonstrate consistent effects across tissues and are involved in immune evasion mechanisms relevant to endometriosis pathogenesis. The reproducibility of these findings across ancestries suggests fundamental immune mechanisms in disease development.
Combinatorial analytics approaches have identified novel gene associations not previously detected through GWAS, providing new insights into autophagy and macrophage biology in endometriosis [2]. These discoveries highlight the value of diverse ancestral representation in uncovering previously overlooked biological mechanisms, potentially offering new targets for therapeutic intervention.
Diagram Title: Cross-ancestry Endometriosis Pathways
Optimizing statistical power in underrepresented ancestral groups requires both methodological innovations and a fundamental shift in research practices. The protocols outlined here provide a framework for enhancing discovery and equity in endometriosis genetics research. Key principles include prioritizing ancestral diversity over mere sample size increases, implementing cross-ancestry validation as a standard practice, and integrating functional genomics to interpret findings across diverse populations.
The field is moving toward approaches that explicitly account for and leverage human genetic diversity, as demonstrated by methods like PhyloFrame that create ancestry-aware disease signatures without requiring ancestry labels in training data [53]. Future directions should include the development of specialized statistical methods for admixed populations, increased investment in diverse biobanks, and standardized reporting of ancestry-specific and trans-ancestry findings. Through these approaches, endometriosis research can achieve both improved scientific understanding and greater equity in precision medicine applications across all ancestral backgrounds.
Trans-ancestry meta-analysis has emerged as a powerful strategy to enhance the resolution of fine-mapping causal variants in genome-wide association studies (GWAS). By leveraging genetic differences across diverse populations, researchers can overcome limitations imposed by linkage disequilibrium (LD) patterns in single-ancestry studies. This Application Note provides detailed protocols for implementing trans-ancestry fine-mapping approaches, with specific application to endometriosis research. We present quantitative comparisons of fine-mapping performance, experimental workflows for cross-population analysis, and essential reagent solutions to facilitate implementation in research and drug discovery settings.
Endometriosis is a heritable hormone-dependent gynecological disorder affecting 6-10% of women of reproductive age, characterized by severe pelvic pain and reduced fertility [58]. Genome-wide association studies have identified numerous loci associated with endometriosis risk, yet identifying precise causal variants remains challenging due to extensive LD in single populations [58] [59].
Trans-ancestry meta-analysis leverages differential LD patterns across populations to improve fine-mapping resolution. When causal variants are shared across populations but tagged by different haplotype structures due to varying LD patterns, combining data from diverse ancestry groups enables more precise identification of causal variants [60] [61]. This approach is particularly valuable for endometriosis research, where previous studies have identified risk loci in genes involved in sex steroid hormone pathways including FN1, CCDC170, ESR1, SYNE1, and FSHB [58].
Table 1: Performance Comparison of Fine-Mapping Approaches in Simulated Data
| Method | Single-Ancestry Credible Set Size | Trans-Ancestry Credible Set Size | Causal Variants Identified (PIP >0.5) | Computational Requirements |
|---|---|---|---|---|
| MESuSiE | 44 (EUR), 21 (EAS) | 54 regions | 25 | High |
| SuSiE | 44 (EUR), 21 (EAS) | - | Fewer than MESuSiE | Moderate |
| MR-MEGA | - | 6 novel loci detected | Improved over fixed-effects | Low |
| Fixed-effect meta-analysis | - | 13 novel loci | Standard approach | Low |
The following diagram illustrates the comprehensive workflow for trans-ancestry fine-mapping in endometriosis research:
Diagram 1: Trans-ancestry fine-mapping workflow. The process begins with collection of GWAS summary statistics from diverse populations, proceeds through quality control and meta-analysis, and culminates in functional validation of identified causal variants.
Purpose: To identify and fine-map endometriosis risk loci through trans-ancestry meta-analysis.
Materials:
Procedure:
Data Collection and Harmonization
Trans-ancestry Meta-analysis
Heterogeneity Assessment
Fine-mapping Implementation
Functional Annotation
The following diagram illustrates the logical framework for identifying causal variants through trans-ancestry approaches:
Diagram 2: Causal variant identification logic. Differential LD patterns across populations help narrow the candidate region, with functional evidence confirming true causal variants.
In endometriosis, previous trans-ancestry analyses have identified 19 independent SNPs robustly associated with disease risk, explaining up to 5.19% of variance [58]. The following table summarizes key endometriosis loci identified through trans-ancestry approaches:
Table 2: Endometriosis Risk Loci Identified Through Trans-ancestry Meta-analysis
| Locus | Gene | Lead SNP | Odds Ratio | P-value | Function |
|---|---|---|---|---|---|
| 6q25.1 | CCDC170 | rs1971256 | 1.09 | 3.74Ã10â»â¸ | Hormone metabolism |
| 6q25.1 | SYNE1 | rs71575922 | 1.11 | 2.02Ã10â»â¸ | Nuclear organization |
| 11p14.1 | FSHB | rs74485684 | 1.11 | 2.00Ã10â»â¸ | Gonadotropin subunit |
| 2q35 | FN1 | rs1250241 | 1.23 | 2.99Ã10â»â¹ | Extracellular matrix |
| 7p12.3 | - | rs74491657 | 1.46 | 4.71Ã10â»â¹ | Unknown |
Table 3: Essential Research Reagents and Tools for Trans-ancestry Fine-Mapping
| Reagent/Tool | Function | Application Notes |
|---|---|---|
| MR-MEGA | Trans-ethnic meta-regression | Accounts for heterogeneity correlated with ancestry [60] |
| MESuSiE | Cross-population fine-mapping | Identifies shared and ancestry-specific causal signals [61] |
| METAL | Fixed-effects meta-analysis | Standard for GWAS meta-analysis [58] [61] |
| ARTP3 | Trans-ancestry pathway analysis | Integrates SNP signals across ancestry groups [63] [39] |
| 1000 Genomes Project | Reference panel | Provides LD information for diverse populations [58] |
| ANNOVAR | Functional variant annotation | Prioritizes variants by functional impact [62] |
| FINEMAP | Bayesian fine-mapping | Computes posterior inclusion probabilities |
Purpose: To identify biological pathways associated with endometriosis through trans-ancestry pathway analysis.
Materials:
Procedure:
Data Integration
Pathway Analysis
Interpretation
Trans-ancestry approaches substantially improve fine-mapping resolution for endometriosis risk loci by leveraging differential LD patterns across populations. Methods such as MR-MEGA and MESuSiE demonstrate superior performance compared to single-ancestry approaches, reducing credible set sizes and increasing the probability of identifying causal variants [60] [61]. For endometriosis research, these approaches have highlighted the importance of genes involved in sex steroid hormone signaling, including ESR1 and FSHB [58].
Implementation requires careful attention to quality control, ancestry representation, and functional validation. Future directions include integrating trans-ancestry fine-mapping with single-cell epigenomics in endometriosis-relevant tissues and developing polygenic risk scores that transfer across populations for improved risk prediction and clinical translation.
Polygenic risk scores (PRS) have emerged as powerful tools in human genetics, quantifying an individual's inherited susceptibility to complex diseases based on the cumulative effect of numerous genetic variants. However, a significant challenge hindering their equitable clinical application is the sharply reduced accuracy of PRS when applied to non-European populations [64] [65]. This performance disparity stems largely from the historical underrepresentation of diverse populations in genome-wide association studies (GWAS), which are the foundation for calculating these scores. Consequently, PRS developed primarily in European cohorts capture patterns of genetic variation and linkage disequilibrium (LD) specific to that ancestry, limiting their transferability [66] [67].
Enhancing the cross-population accuracy of PRS is not merely a technical statistical challenge but a critical imperative for global health equity. This document details advanced methodologies and protocols for improving PRS transferability, framed within the specific context of endometriosis research. Endometriosis, a common gynecological condition affecting ~10% of women, has a substantial genetic component, but its genetic architecture has been predominantly studied in populations of European ancestry [19] [2]. We focus on trans-ancestry meta-analysis approaches that leverage genetic data from diverse populations to build more portable and powerful risk prediction models.
The reduced portability of PRS across populations is attributed to several key factors:
Table 1: Key Factors Limiting PRS Transferability and Their Consequences.
| Factor | Description | Impact on PRS Accuracy |
|---|---|---|
| LD Structure Variation | Differences in correlation patterns between genetic variants across populations. | Effect sizes from a source population poorly tag causal variants in the target population. |
| Allele Frequency Divergence | Varying frequencies of risk alleles across ancestral groups. | Reduces variance explained by the PRS and fails to capture population-specific risk. |
| Varying Causal Effects | True biological effect of a variant may differ across ancestries. | Introduces systematic bias in risk prediction if not accounted for. |
| Limited Diversity in GWAS | Over-reliance on European-ancestry discovery cohorts. | Fundamental data limitation; models are not trained to recognize risk variants in other groups. |
Several sophisticated statistical methods have been developed to address the limitations of PRS transferability. These approaches can be broadly categorized into those that leverage multi-ancestry GWAS summary statistics and those that employ novel modeling techniques.
Integrating genetic data from multiple populations during the discovery phase is a foundational strategy. A multi-ancestry meta-analysis for endometriosis, encompassing over 1.4 million women (including 105,869 cases), has identified 80 genome-wide significant loci, 37 of which are novel [13]. This expanded genetic map across ancestries provides a more robust set of variants for PRS construction.
Simply pooling data is insufficient; methods must explicitly account for inter-ancestry differences.
Table 2: Comparison of Advanced Statistical Methods for Cross-Population PRS.
| Method | Core Principle | Key Inputs | Reported Improvement |
|---|---|---|---|
| SDPRX | Models joint effect size distribution across populations; auto-adjusts for LD. | GWAS summary stats from two populations. | Improved accuracy over existing methods in non-European populations via simulations and real traits [64]. |
| PolyPred/PolyPred+ | Combines fine-mapping-based causal effect estimates with standard PRS; can integrate target population data. | GWAS summary stats; (Optional) Target population genotype data. | +7% to +32% relative improvement vs. BOLT-LMM in Africans and South Asians; +24% in East Asians with PolyPred+ [65]. |
| PRS-CSx | Bayesian modeling with a shared continuous shrinkage prior across multiple populations. | GWAS summary stats from multiple populations; Population-matched LD reference panels. | Effective for T2D prediction in trans-ancestry cohorts (European, African, Hispanic/Latino) [67]. |
| GPSMult | Integrates cross-ancestry GWAS for the primary trait and multiple genetically correlated risk factors. | Large-scale GWAS summary stats for a disease and its risk factors across ancestries. | Outperformed all previously published CAD PRS in multi-ethnic validation datasets [66]. |
Moving beyond common variant-based PRS, novel analytical frameworks are emerging.
This section provides a detailed, actionable protocol for developing and validating a trans-ancestry PRS for endometriosis.
Objective: To develop a polygenic risk score for endometriosis with improved predictive accuracy across diverse populations by integrating multi-ancestry GWAS summary statistics using the PRS-CSx method.
Diagram 1: Trans-ancestry PRS development workflow.
Table 3: Research Reagent Solutions for Trans-ancestry PRS Analysis.
| Item | Function/Description | Example Sources |
|---|---|---|
| GWAS Summary Statistics | Effect sizes, standard errors, and p-values for genetic variants associated with endometriosis. | International Endogene Consortium [19], FinnGen, Biobank Japan, All of Us [13]. |
| LD Reference Panels | Genotype data used to estimate population-specific linkage disequilibrium patterns. | 1000 Genomes Project (1KG) [67], HRC, ancestry-specific reference panels. |
| Genotyped Target Cohorts | Independent datasets with individual-level genotype and phenotype data for validation. | UK Biobank, All of Us, Taiwan Biobank, etc. [67] [13]. |
| High-Performance Computing (HPC) Cluster | Essential for running computationally intensive genetic analyses and Bayesian methods. | Local institutional HPC or cloud computing services (e.g., AWS, Google Cloud). |
| Analysis Software & Packages | Specialized tools for PRS construction and analysis. | PRS-CSx [67], SDPRX [64], PolyFun/PolyPred [65], PLINK, R/Bioconductor. |
Data Collection and Curation
Data Quality Control and Harmonization
PRS Construction using PRS-CSx
python PRS-CSx.py --ref_dir=[PATH_TO_LD] --bim_prefix=[TARGET_BIM] --sst_file=[EUR_SUMSTATS],[NON_EUR_SUMSTATS] --n_gwas=[EUR_N],[NON_EUR_N] --out_dir=[OUTPUT_DIR]--phi parameter can be set to auto for automatic learning of the shrinkage parameter. Specify the population labels for each set of summary statistics.PRS Calculation and Validation
Biological Interpretation and Downstream Analysis
Objective: To experimentally validate a candidate protein target (e.g., RSPO3) identified through trans-ancestry PRS and downstream MR analysis in clinical endometriosis samples [6].
Diagram 2: Experimental validation workflow for candidate targets.
The equitable application of polygenic risk scores in clinical practice hinges on our ability to improve their accuracy across the full spectrum of human genetic diversity. Methods such as SDPRX, PolyPred/PolyPred+, and PRS-CSx provide powerful statistical frameworks for achieving this by explicitly modeling ancestral differences and leveraging diverse data. Within endometriosis research, the ongoing generation of large-scale multi-ancestry GWAS data [13], combined with these advanced methods and subsequent experimental validation [6], creates a transformative pathway. This integrated approach promises not only to improve risk prediction for all women but also to uncover novel biology and therapeutic targets, ultimately advancing precision medicine for this common and debilitating condition.
Within the specific context of endometriosis genetics research, a disease with complex heritability and significant diagnostic challenges, the need for precise genetic risk prediction is paramount [8] [19]. Genome-wide association studies (GWAS) have successfully identified multiple loci associated with endometriosis risk [21]. However, the predominant reliance on European-ancestry cohorts has limited the generalizability of resulting polygenic risk scores (PRS) across diverse populations, a critical issue for global drug development and clinical application [68].
Trans-ancestry meta-analysis methods present a promising solution to mitigate these biases. These approaches leverage genetic data from multiple populations to enhance the discovery of risk loci and improve the portability of PRS [61]. This protocol details a comprehensive framework for benchmarking trans-ancestry PRS models against ancestry-specific alternatives in endometriosis research, providing drug development professionals with standardized methods for evaluating genetic risk prediction tools.
PRS quantify an individual's genetic susceptibility to a trait by aggregating the effects of numerous genetic variants, typically identified through GWAS [69]. Traditional PRS methods, such as clumping and thresholding (C+T), often demonstrate reduced predictive accuracy when applied to populations not represented in the original training GWAS, particularly for complex diseases like endometriosis [68] [69].
Endometriosis is a heritable, estrogen-dependent inflammatory disease affecting approximately 6-10% of women of reproductive age [19] [22]. Its genetic architecture is complex, with a common SNP-based heritability estimated at 0.26 [21]. Large-scale meta-analyses have identified numerous susceptibility loci, many implicating genes involved in sex steroid hormone pathways (e.g., WNT4, VEZT, ESR1, FSHB), highlighting potential therapeutic targets [21].
Table 1: Key Endometriosis Susceptibility Loci from GWAS Meta-Analyses
| Locus | Nearest Gene | Reported Function | Population | P-value | Reference |
|---|---|---|---|---|---|
| 7p15.2 | - | Inter-genic | European | 1.6 à 10â»â¹ | [19] |
| 1p36.12 | WNT4 | Developmental pathways | European | 1.8 à 10â»Â¹âµ | [19] [21] |
| 12q22 | VEZT | Cell adhesion | European | 4.7 à 10â»Â¹âµ | [19] |
| 9p21.3 | CDKN2B-AS1 | Cell cycle regulation | Japanese | 5.57 à 10â»Â¹Â² | [19] |
| 6q25.1 | CCDC170/ESR1 | Hormone metabolism | Trans-ancestry | 3.74 à 10â»â¸ | [21] |
| 11p14.1 | FSHB | Hormone metabolism | Trans-ancestry | 2.00 à 10â»â¸ | [21] |
This section outlines the core experimental workflow for benchmarking PRS models, from data preparation through to performance evaluation. The following diagram illustrates the complete process:
GWAS Summary Statistics
Linkage Disequilibrium (LD) Reference Panels
Ancestry-Specific PRS
Trans-Ancestry PRS
Parameter Optimization
Benchmarking Cohorts
Evaluate PRS performance using multiple statistical measures to ensure comprehensive assessment. The following diagram illustrates the relationship between evaluation components:
Primary Metrics
Table 2: Example PRS Performance Comparison for Complex Traits (Adapted from Published Studies)
| Trait | Population | Ancestry-Specific PRS (R²) | Trans-Ancestry PRS (R²) | European PRS (R²) | Effect Size Contrast | Reference |
|---|---|---|---|---|---|---|
| LDL Cholesterol | East Asian (TWB) | 9.3% | 6.7% | 4.5% | 0.82 vs 0.76 vs 0.59* | [68] |
| LDL Cholesterol | East Asian (UKB) | 8.6% | 7.8% | 6.2% | - | [68] |
| Kidney Stone Disease | Trans-ancestry | - | PRS-CSxEAS&EUR (Superior) | - | OR: 1.83 (1.68-1.98) | [61] |
| *Mean difference in LDL levels between extreme PRS deciles | ||||||
| OR for highest vs. middle PRS quintile |
The scPRS framework represents a cutting-edge advancement that integrates single-cell epigenomics with PRS calculation [69]. This approach is particularly relevant for endometriosis given its tissue-specific pathophysiology.
Workflow Implementation
Trans-ancestry Mendelian randomization can elucidate potential causal relationships between modifiable risk factors and endometriosis [22]. For instance, a recent trans-ethnic MR study investigated the causal effects of serum lipids on endometriosis risk, identifying triglyceride-lowering gene targets (LPL, PPARA, ANGPTL3, APOC3) as potential therapeutic avenues [22].
Table 3: Essential Research Reagents and Computational Tools
| Category | Item | Function/Application | Example Resources |
|---|---|---|---|
| Data Sources | GWAS Summary Statistics | Effect size estimates for variants | GWAS Catalog (EFO_0001065), IEC, BBJ [8] [19] |
| LD Reference Panels | Population-specific linkage patterns | 1000 Genomes Project, UK Biobank, TWB [68] [61] | |
| Software Tools | PRS-CSx | Trans-ancestry PRS construction | GitHub: getian107/PRScsx [68] |
| PUMAS/PUMAS-ensemble | Summary-statistics-based tuning & ensemble learning | [70] | |
| scPRS | Single-cell PRS calculation | [69] | |
| TwoSampleMR | Mendelian randomization analysis | [22] | |
| Biobanks | Taiwan Biobank (TWB) | East Asian validation cohort | [68] |
| UK Biobank (UKB) | European validation cohort | [68] [70] | |
| Biobank Japan (BBJ) | East Asian GWAS data | [61] |
Benchmarking trans-ancestry PRS against ancestry-specific models represents a critical methodological advancement in endometriosis genetics. The protocols outlined here provide a rigorous framework for evaluating PRS performance across diverse populations, directly addressing the critical need for portable genetic risk tools in global drug development programs. As trans-ancestry resources expand, these approaches will become increasingly integral to identifying valid therapeutic targets and developing stratified treatment strategies for endometriosis and other complex genetic disorders.
This application note details a combinatorial analytics approach for identifying and validating multi-Single Nucleotide Polymorphism (SNP) signatures in endometriosis research. Conventional genome-wide association studies (GWAS) have explained only approximately 5% of disease variance in endometriosis, revealing the need for more sophisticated analytical methods to capture its complex genetic architecture [2]. Combinatorial analytics addresses this limitation by detecting synergistic effects between multiple genetic variants that are undetectable through single-variant analysis.
The protocol outlined herein enabled the identification of 1,709 reproducible disease signatures comprising 2,957 unique SNPs in combinations of 2-5 SNPs, demonstrating significant enrichment (58-88%) across diverse ancestry cohorts [2]. This approach has revealed novel biological pathways and potential therapeutic targets, moving beyond the constraints of traditional GWAS and providing a framework for precision medicine in endometriosis and other complex disorders.
Endometriosis affects approximately 10% of women of reproductive age globally, yet diagnosis is typically delayed by 7-10 years due to limited understanding of its pathogenesis and lack of non-invasive diagnostic tools [2] [49]. While familial aggregation and twin studies provide evidence of a strong heritable component, traditional GWAS approaches have identified only 42 genomic loci associated with endometriosis risk, collectively explaining just 5% of disease variance [2].
Combinatorial analytics represents a paradigm shift by analyzing how multiple SNPs interact to influence disease risk, potentially capturing non-linear genetic effects that single-variant approaches miss. This method aligns with broader efforts in trans-ancestry genetic research, which aims to improve the generalizability of findings across diverse populations and enhance the biological relevance of discovered associations [49] [71].
Objective: Identify statistically significant multi-SNP combinations associated with endometriosis risk.
Materials:
Procedure:
Output: 1,709 disease signatures comprising 2,957 unique SNPs.
Objective: Validate discovered signatures in diverse genetic backgrounds.
Materials:
Procedure:
Output: Reproducibility metrics across ancestry groups and signature frequency categories.
Objective: Interpret validated signatures in functional biological context.
Materials:
Procedure:
Output: Annotated list of biological pathways and prioritized therapeutic targets.
Table 1: Reproduction Rates of Multi-SNP Signatures by Population and Frequency
| Signature Frequency | All of Us Overall | Non-European Cohorts | Key Findings |
|---|---|---|---|
| >9% (High Frequency) | 80-88% (p<0.01) | 66-76% (p<0.04) | Highest reproducibility across all groups |
| 4-9% (Medium Frequency) | 70-78% (p<0.03) | 60-70% (p<0.04) | Moderate but significant reproduction |
| <4% (Low Frequency) | 58-65% (p<0.04) | 55-62% (p<0.05) | Lowest but above-chance reproduction |
The validation demonstrated exceptionally high reproducibility for frequent signatures, with one 2-SNP signature achieving individual significance in the AoU cohort [2]. Notably, reproducibility remained substantial even in non-European populations, supporting the trans-ancestry robustness of the combinatorial approach.
Table 2: Gene Categories Identified Through Combinatorial Analysis
| Gene Category | Count | Examples | Biological Significance |
|---|---|---|---|
| Previously Established GWAS Genes | 7 | Known from meta-GWAS | Validation of existing findings |
| Literature-Associated with Endometriosis | 16 | Documented in OpenTargets | Confirmation of prior evidence |
| Novel Gene Associations | 75 | MAP3K5, autophagy and macrophage genes | New biological mechanisms |
| High-Priority Therapeutic Targets | 9 | Characterized novel genes | Credible drug discovery/repurposing candidates |
The combinatorial method identified 75 novel gene associations not detected by previous GWAS, significantly expanding the known genetic landscape of endometriosis [2]. Several of these novel genes implicate previously underappreciated biological processes in endometriosis, including autophagy and macrophage biology.
The pathway analysis revealed enrichment in several biologically relevant processes. Cell adhesion, proliferation, and migration pathways align with the invasive nature of endometriotic lesions. Cytoskeleton remodeling and angiogenesis are essential for lesion establishment and maintenance. The novel associations with autophagy and macrophage biology suggest involvement of cellular clearance mechanisms and immune microenvironment modulation in disease pathogenesis [2] [10].
Integration with multi-omic data through summary-based Mendelian randomization (SMR) approaches has further validated these pathways, demonstrating coordinated effects across methylation, gene expression, and protein abundance layers [10].
Table 3: Essential Research Materials and Platforms for Combinatorial Analytics
| Resource Category | Specific Solutions | Application in Workflow |
|---|---|---|
| Analytical Platforms | PrecisionLife combinatorial analytics platform | Primary analysis of multi-SNP combinations |
| Genetic Datasets | UK Biobank, All of Us Research Program, FinnGen | Discovery and validation cohorts |
| Bioinformatics Tools | SMR software (v1.3.1), PRS-CSx, LOG-TRAM | Multi-omic integration and trans-ancestry optimization |
| Pathway Databases | CellAge, KEGG, Reactome, GO, GTEx | Biological interpretation and functional annotation |
| Quality Control Tools | PLINK, R/bioconductor packages | Data preprocessing and population structure control |
The PrecisionLife platform served as the core analytical engine, specifically designed to detect combinatorial effects in complex disease datasets [2]. The integration of large-scale biobanks provided both discovery (UK Biobank) and validation (All of Us) cohorts with sufficient sample size and ancestral diversity. Specialized statistical genetics tools like PRS-CSx and LOG-TRAM enabled effective cross-population analysis by accounting for ancestry-specific linkage disequilibrium patterns [67] [71].
Combinatorial analytics represents a powerful approach for unraveling the complex genetic architecture of endometriosis, significantly outperforming traditional GWAS in both novel biological discovery and cross-population validation. The successful identification of 75 novel gene associations and the high reproducibility rates (up to 88%) across diverse ancestry groups demonstrates the method's robustness and translational potential [2].
This protocol provides researchers with a comprehensive framework for implementing combinatorial analytics in complex trait genetics. The integration of these methods with trans-ancestry meta-analysis approaches promises to accelerate the discovery of biologically relevant therapeutic targets and advance precision medicine for endometriosis and other complex disorders with similarly elusive genetic architectures.
Endometriosis is a heritable, estrogen-dependent gynecological disorder affecting approximately 6-10% of women of reproductive age, characterized by the presence of endometrial-like tissue outside the uterus and associated with chronic pelvic pain and reduced fertility [21] [19]. The genetic architecture of endometriosis involves multiple loci with modest effects, and trans-ancestry meta-analysis approaches have proven invaluable for disentangling this complexity by leveraging genetic differences across populations. Cross-population colocalization and fine-mapping represent advanced statistical genetic methodologies that enable more precise identification of causal variants and genes by integrating genome-wide association data (GWAS) from diverse ancestral backgrounds. These approaches effectively address the limitations of single-ancestry studies by leveraging differences in linkage disequilibrium patterns and allele frequencies across populations, leading to refined candidate causal variants and enhanced biological insights for therapeutic development [12] [13].
The implementation of these methods in recent large-scale genetic studies of endometriosis has dramatically expanded our understanding of its genetic underpinnings, revealing novel risk loci and highlighting the critical role of sex steroid hormone pathways, immune regulation, and tissue remodeling mechanisms in disease pathogenesis [21] [12]. This application note provides a comprehensive framework for implementing cross-population colocalization and fine-mapping validation within the context of endometriosis research, including standardized protocols, data resources, and analytical workflows to accelerate gene discovery and drug target prioritization.
Table 1: Summary of Key Endometriosis Genetic Association Studies
| Study | Sample Size (Cases/Controls) | Ancestries | Significant Loci | Novel Loci | Primary Findings |
|---|---|---|---|---|---|
| Nyholt et al. 2012 [48] | 4,604/9,393 | European, Japanese | 7 | 4 | First trans-ancestry meta-analysis; identified WNT4, GREB1, VEZT loci; demonstrated shared genetic architecture |
| Sapkota et al. 2017 [21] | 17,045/191,596 | European, Japanese | 19 (9 replicated + 5 novel + 5 secondary) | 5 | Highlighted key genes in hormone metabolism (FN1, CCDC170, ESR1, SYNE1, FSHB); explained up to 5.19% variance |
| Multi-ancestry 2025 (Preprint) [12] [13] | ~105,869/~1.3 million | Multi-ancestry | 80 | 37 | Largest study to date; first adenomyosis loci; identified pathways for immune regulation, tissue remodeling, cell differentiation |
| FinnGen R10 [10] | 16,588/111,583 | European | N/A | N/A | Validation cohort for cell aging genes; confirmed THRB and ENG protein associations |
Table 2: Essential Data Resources for Cross-Population Analysis
| Data Type | Source | Sample Size | Ancestries | Application in Endometriosis Research |
|---|---|---|---|---|
| GWAS Summary Statistics | FinnGen R10 [10] | 16,588 cases/111,583 controls | European | Primary discovery and validation |
| UK Biobank [11] [10] | 4,036 cases/210,927 controls | European | Replication and meta-analysis | |
| BioBank Japan [21] [48] | 1,423 cases/1,318 controls | Japanese | Trans-ancestry discovery | |
| Expression QTLs (eQTLs) | eQTLGen [72] [10] | 31,684 individuals | Mostly European | Blood-based gene expression regulation |
| GTEx v8 [72] [10] | 838 donors (17,382 samples) | Multi-ancestry | Tissue-specific regulation (including uterus) | |
| Methylation QTLs (mQTLs) | BSGS/LBC meta-analysis [10] | 1,980 individuals | European | Epigenetic regulation in blood |
| Protein QTLs (pQTLs) | UK Biobank Pharma Proteomics [10] | 54,219 participants | European | Plasma protein abundance regulation |
| Iceland pQTL [11] | 35,559 individuals | European | Independent pQTL validation |
Purpose: To refine causal variant identification by leveraging differential linkage disequilibrium patterns across diverse populations.
Workflow:
Data Harmonization
Conditional Analysis
Fine-Mapping Implementation
Cross-Population Integration
Figure 1: Cross-population fine-mapping workflow for identifying causal variants by leveraging differential linkage disequilibrium across diverse ancestral groups.
Purpose: To identify shared genetic signals between endometriosis risk and molecular quantitative trait loci, providing evidence for potential causal mechanisms.
Workflow:
Variant Selection
Colocalization Testing
coloc R packageSensitivity Analyses
Multi-omic Integration
Figure 2: Multi-omic colocalization analysis workflow for identifying shared genetic signals between endometriosis risk and molecular quantitative trait loci across diverse data types.
Purpose: To prioritize and validate potential drug targets for endometriosis using genetic evidence across populations.
Workflow:
Target Identification
Causal Evidence Assessment
Cross-Ancestry Replication
Therapeutic Potential Evaluation
Table 3: Key Research Reagents and Computational Resources
| Category | Resource/Tool | Application | Key Features |
|---|---|---|---|
| GWAS Data | FinnGen R10 [10] | Endometriosis genetic associations | 16,588 cases, 111,583 controls; European ancestry |
| UK Biobank [11] | Validation cohort | 4,036 cases, 210,927 controls; deep phenotyping | |
| BBJ [21] | Trans-ancestry discovery | Japanese ancestry; enables cross-population analysis | |
| QTL Resources | GTEx v8 [72] [10] | Tissue-specific eQTLs | Uterus and 51 other tissues; multi-ancestry |
| eQTLGen [72] [10] | Blood eQTLs | 31,684 individuals; largest blood eQTL resource | |
| Iceland pQTL [11] | Plasma protein QTLs | 4,907 cis-pQTLs; SOMAscan platform | |
| Software Tools | GCTA-COJO | Conditional analysis | Identifies independent association signals |
| SUSIE/FINEMAP | Fine-mapping | Bayesian methods for credible set construction | |
| coloc R package [72] [10] | Colocalization analysis | Bayesian test for shared causal variants | |
| SMR [10] | Mendelian randomization | Integrates GWAS and QTL data for causal inference | |
| Experimental Validation | ELISA kits [11] | Protein quantification | Validate pQTL findings (e.g., RSPO3 levels) |
| RT-qPCR [11] | Gene expression | Confirm differential expression in tissues | |
| Clinical samples [11] | Functional validation | Endometriosis lesions vs. control endometrium |
A recent study demonstrated the application of these protocols by identifying RSPO3 (R-spondin 3) as a potential therapeutic target for endometriosis [11]. The researchers:
This multi-step approach exemplifies how cross-population colocalization and validation can prioritize high-confidence drug targets with translational potential.
Another application utilized multi-omic SMR analysis to investigate cell aging-related genes in endometriosis pathogenesis [10]. The study:
Ancestral Diversity Limitations: When working with under-represented populations, consider using trans-ancestry fine-mapping methods that can handle sparse data.
Sample Overlap: Account for potential sample overlap between GWAS and QTL studies using appropriate correlation matrices.
Power Considerations: Ensure adequate sample sizes for colocalization analysis, particularly for cell-type specific QTLs with smaller effect sizes.
Multiple Testing: Apply conservative significance thresholds (e.g., Bonferroni correction) when testing multiple genes or genomic regions.
Functional Interpretation: Integrate epigenomic annotation (e.g., ENCODE, Roadmap) to prioritize variants with regulatory potential.
These protocols provide a standardized framework for implementing cross-population colocalization and fine-mapping in endometriosis research, enabling more robust gene discovery and therapeutic target identification across diverse ancestral groups.
Within the context of advancing trans-ancestry meta-analysis methods for endometriosis genome-wide association studies (GWAS), a critical challenge remains: translating genetic discoveries into clinically actionable insights for diverse patient populations. Endometriosis is a common chronic condition affecting approximately 10% of women of reproductive age, characterized by debilitating symptoms including chronic pelvic pain and fatigue, yet it suffers from diagnostic delays of 7-9 years on average and limited treatment options [73] [2]. The heterogenous symptom presentation and substantial comorbidity profile observed in endometriosis patients complicate clinical management and therapeutic development. This Application Note provides detailed protocols for assessing the predictive utility of novel genetic and digital biomarkers for specific symptom subtypes and comorbidities, facilitating their translation into clinical applications and precision medicine approaches.
Traditional GWAS approaches in endometriosis have identified multiple genomic loci associated with disease risk, but these explain only approximately 5% of disease variance [2]. This limitation highlights the complex genetic architecture of endometriosis and the need for more sophisticated analytical methods. Trans-ancestry meta-analysis offers enhanced power for locus discovery and fine-mapping, potentially revealing population-specific and shared genetic risk factors. However, the clinical utility of these findings depends on robust validation and connection to phenotypic manifestations.
Recent research demonstrates that combinatorial analytics and Mendelian randomization (MR) approaches can identify novel genetic risk factors and potential therapeutic targets that may be overlooked by conventional GWAS [2] [6]. Simultaneously, digital technologies including actigraphy and smartphone-based monitoring provide objective measures of symptoms and behaviors that correlate with patient-reported outcomes, offering new avenues for quantifying symptom severity and trajectory [73] [74]. Integrating these multidimensional data sources is essential for developing comprehensive biomarkers capable of predicting symptom subtypes and comorbidities.
Table 1: Key Genetic Findings from Combinatorial Analytics in Endometriosis
| Analysis Type | Cohort | Key Findings | Clinical Translation Potential |
|---|---|---|---|
| Combinatorial Analytics [2] | UK Biobank (White European), All of Us (Multi-ancestry) | 1,709 disease signatures identified; 58-88% reproducibility in validation cohort; 75 novel genes discovered | Pathways identified (cell adhesion, fibrosis, neuropathic pain) inform subtype stratification; novel therapeutic targets |
| Mendelian Randomization [6] | UK Biobank, FinnGen | RSPO3 and FLT1 potentially associated with endometriosis; robust association confirmed for RSPO3 | Causal evidence supporting RSPO3 as therapeutic target; potential biomarker for patient stratification |
| Trans-ancestry Reproducibility [2] | All of Us sub-cohorts | 66-76% reproducibility of signatures in non-white European cohorts | Demonstrates utility of genetic signatures across ancestries; supports inclusive biomarker development |
Table 2: Digital Biomarker Correlations with Endometriosis Symptoms
| Digital Measure | Symptom Correlation | Study Details | Clinical Utility |
|---|---|---|---|
| Physical Activity (Actigraphy) [74] | Strong negative correlation with fatigue (R < -0.3) | 68 participants, up to three 4-6 week monitoring cycles | Objective measure of fatigue impact; treatment monitoring |
| Activity Rhythms & Sleep [74] | Associated with symptom severity and variability (â®Râ® > 0.3) | 5152 days of actigraphy data | Identifies patients with more severe symptom trajectories |
| Post-surgical Changes [74] | Reflect changes in self-reported symptoms | n=16 surgical patients | Objective measure of treatment response |
Purpose: To validate novel combinatorial genetic signatures identified through trans-ancestry meta-analysis in diverse patient populations.
Materials:
Procedure:
Analysis:
Purpose: To objectively characterize symptom trajectories and treatment responses using wearable device data.
Materials:
Procedure:
Analysis:
Purpose: To provide causal evidence for potential therapeutic targets and validate them in clinical samples.
Materials:
Procedure:
Analysis:
Table 3: Essential Research Reagents and Platforms for Endometriosis Biomarker Studies
| Category | Specific Products/Platforms | Function | Key Considerations |
|---|---|---|---|
| Genetic Analysis | PrecisionLife combinatorial analytics platform [2], PLINK, METAL | Identify and validate genetic risk signatures beyond GWAS | Handles high-order SNP interactions; validated in trans-ancestry cohorts |
| Wearable Sensors | ActiGraph, Fitbit, Apple Watch [73] [74] | Continuous collection of actigraphy data for digital phenotyping | Balance between research-grade accuracy and patient acceptability |
| Biomolecular Assays | Human R-Spondin3 ELISA Kit [6], SOMAscan, RNA extraction kits | Quantify protein and gene expression biomarkers | Sensitivity and specificity for low-abundance targets; validation in relevant matrices |
| Data Integration | R (urbnthemes package) [75], Python pandas, SQL databases | Manage and analyze multi-modal data streams | Ensure interoperability between genetic, digital, and clinical data |
| Statistical Analysis | Mendelian randomization packages (TwoSampleMR, MRBase) [6], mixed-effects models | Establish causal relationships and longitudinal patterns | Account for population structure and repeated measures |
The integration of trans-ancestry genetic data with multidimensional phenotypic information from digital health technologies creates unprecedented opportunities for advancing precision medicine in endometriosis. The protocols outlined in this Application Note provide a roadmap for robust validation of biomarkers capable of predicting symptom subtypes and comorbidities, ultimately facilitating targeted therapeutic development and personalized management approaches. As these methods continue to evolve, they hold promise for reducing the diagnostic delay and improving quality of life for the diverse population of individuals affected by endometriosis.
Trans-ancestry genetic association studies are essential for identifying robust and generalizable genetic risk factors for endometriosis. Two primary analytical approachesâmeta-analysis and mega-analysisâenable the combination of genetic data across diverse ancestral populations. This application note provides a structured comparison of these methodologies, detailing protocols for their implementation in endometriosis research. We present quantitative performance metrics, experimental workflows, and essential research tools to guide researchers in selecting and executing the optimal analytical strategy for their trans-ancestry studies.
Endometriosis is a complex gynecological disorder affecting approximately 10% of women of reproductive age, with a significant genetic component contributing to its pathogenesis [8]. Genome-wide association studies (GWAS) have successfully identified multiple susceptibility loci for endometriosis; however, many early studies were limited by a predominant focus on European-ancestry populations [19] [21]. Trans-ancestry genetic studies are critically important for improving the power and generalizability of findings, facilitating fine-mapping of causal variants, and enhancing equity in genetic research [36].
The integration of diverse ancestry groups in genetic studies presents methodological challenges, particularly in selecting the optimal approach for combining genetic data. Meta-analysis, which combines summary statistics from analyses performed within homogeneous ancestry groups, has been the traditional approach for multi-ancestry GWAS [36]. More recently, mega-analysisâwhich pools individual-level data across ancestry groups for unified processing and analysisâhas gained traction with the emergence of cosmopolitan reference panels like TOPMed [36]. This application note systematically compares these two approaches in the context of endometriosis research, providing detailed protocols and performance metrics to guide researchers in trans-ancestry study design.
Meta-analysis follows a "analyze-first, combine-later" paradigm where genetic data from different ancestry groups are processed and analyzed separately using ancestry-specific reference panels, with summary statistics subsequently combined using fixed or random-effects models [36]. This approach effectively controls for population stratification within homogeneous groups but may exclude individuals of admixed ancestry and reduce statistical power when heterogeneity exists between groups [36].
Mega-analysis employs a "combine-first, analyze-later" approach where individual-level data from all participants are collectively processed using a cosmopolitan reference panel and analyzed in a unified framework that incorporates genetic ancestry as covariates [36]. This integrated approach maximizes sample size and leverages cosmopolitan reference panels but requires careful handling of population stratification across diverse groups [36].
Table 1: Fundamental Methodological Differences Between Meta-Analysis and Mega-Analysis
| Feature | Meta-Analysis | Mega-Analysis |
|---|---|---|
| Data Structure | Summary statistics | Individual-level data |
| Reference Panels | Ancestry-specific (e.g., CAAPA, HRC, GAsP) | Cosmopolitan (e.g., TOPMed) |
| Ancestry Handling | Analysis within homogeneous groups | Unified analysis with ancestry covariates |
| Implementation | Distributed analysis with results combination | Centralized processing and analysis |
| Admixed Individuals | Often excluded | Included with appropriate modeling |
Empirical comparisons in multi-ancestry studies demonstrate distinct performance characteristics for each approach. In a multi-national study of maternal glycemia, mega-analysis identified significantly more genome-wide significant associations compared to meta-analysis, including biologically credible associations at the MTNR1B locus that were not detected by meta-analysis [36]. For metabolomics analyses, the number of significant findings in heterogeneous ancestry mega-analysis "far exceeded" those from homogeneous ancestry meta-analysis and confirmed many previously documented associations [36].
Table 2: Empirical Performance Comparison from Multi-Ancestry Studies
| Performance Metric | Meta-Analysis | Mega-Analysis |
|---|---|---|
| Number of Significant Loci | 15 variants near GCK with maternal fasting glucose [36] | Rich set of variants including GCK and MTNR1B with both fasting and 1-hour glucose [36] |
| Metabolomics Associations | Limited significant findings [36] | Vastly more significant findings [36] |
| Genomic Control | Well-controlled genomic inflation factors [36] | Variable genomic inflation factors requiring careful interpretation [36] |
| Analytical Flexibility | Accommodates study-specific covariates and ancestry adjustments [76] | Enables uniform quality control and analysis framework [36] |
| Implementation Practicality | No individual-level data sharing; efficient for consortia [76] | Requires data harmonization and significant computational resources [36] |
For analysis of gene-environment (GÃE) interactions in trans-ancestry settings, both methods have demonstrated comparable performance. An empirical comparison of four studies found that meta-analysis and mega-analysis provided similar effect size estimates, standard errors, and p-values for GÃE interactions, with highly correlated results (Pearson's r = 0.98) and comparable genomic inflation factors [76].
Principle: Perform GWAS within homogeneous ancestry groups followed by statistical combination of results [19] [21].
Step-by-Step Procedure:
Ancestry Group Definition
Ancestry-Specific Genotype Imputation
Ancestry-Stratified Association Analysis
Summary Statistics Meta-Analysis
Visualization 1: Trans-Ancestry Meta-Analysis Workflow. This diagram illustrates the sequential process of analyzing genetically similar groups separately followed by statistical combination of results.
Principle: Collective processing and unified analysis of individual-level data across diverse ancestry groups [36].
Step-by-Step Procedure:
Cross-Ancestry Genotype Harmonization
Cosmopolitan Reference Panel Imputation
Population Structure Adjustment
Unified Association Analysis
Visualization 2: Trans-Ancestry Mega-Analysis Workflow. This diagram illustrates the integrated approach of combining diverse data before processing and analysis.
Table 3: Key Analytical Tools and Resources for Trans-Ancestry Endometriosis GWAS
| Resource Category | Specific Tool/Resource | Application in Endometriosis Research |
|---|---|---|
| Reference Panels | TOPMed Freeze 8 [36] | Cosmopolitan imputation for mega-analysis |
| CAAPA Panel [36] | African-ancestry specific imputation for meta-analysis | |
| 1000 Genomes Phase 3 [21] | Multi-ancestry reference panel | |
| Imputation Tools | Minimac4 [36] | Efficient genotype imputation |
| Eagle v2.4 [36] | Accurate haplotype phasing | |
| Association Software | METAL [76] | Summary statistics meta-analysis |
| REGENIE [36] | Unified association testing for mega-analysis | |
| Functional Validation | GTEx eQTL Database [8] [5] | Tissue-specific regulatory effect mapping for endometriosis risk loci |
| Ensembl VEP [8] [5] | Functional annotation of endometriosis-associated variants | |
| Data Sources | GWAS Catalog [8] [5] | Repository of published endometriosis associations |
| UK Biobank [6] | Large-scale genetic and phenotypic data | |
| FinnGen [6] | Finnish population cohort with endometriosis cases |
Endometriosis presents unique challenges for genetic analysis due to its heterogeneous clinical presentation, with different genetic effects observed across disease stages. Multiple studies have demonstrated that most endometriosis risk loci show stronger genetic effects for revised American Fertility Society (rAFS) Stage III/IV disease compared to all cases combined [19] [21]. This has important implications for trans-ancestry studies:
The choice between meta-analysis and mega-analysis depends on research objectives, data availability, and computational resources:
Both meta-analysis and mega-analysis offer viable approaches for trans-ancestry endometriosis GWAS, with complementary strengths and limitations. Meta-analysis provides a practical framework for consortium-based research with distributed data, while mega-analysis offers increased power and more unified analytical approaches. The emergence of cosmopolitan reference panels and improved methods for controlling population stratification continue to enhance both approaches. For endometriosis research specifically, attention to disease heterogeneity and integration with functional genomic data from relevant tissues will be essential for translating genetic discoveries into biological insights and therapeutic targets.
Trans-ancestry meta-analysis methods have fundamentally transformed endometriosis genetics, expanding the catalog of risk loci from approximately 45 to over 80 significant associations and providing unprecedented insights into disease biology across diverse populations. The integration of multi-ancestry datasets has enhanced discovery power, improved fine-mapping resolution, and revealed both shared and population-specific risk mechanisms. Methodological advances in polygenic risk scoring, pathway analysis, and multi-omics integration are paving the way for more equitable precision medicine approaches. Future directions should focus on increasing representation of underrepresented populations, developing standardized frameworks for cross-ancestry analysis, and translating genetic discoveries into clinically actionable insights through drug repurposing and targeted therapeutic development. These approaches will ultimately enable earlier diagnosis, improved risk stratification, and more effective, personalized treatments for endometriosis patients worldwide.