OBFC1 is a 368-amino acid protein with two primary structural domains:
N-terminal OB fold: Facilitates single-stranded DNA (ssDNA) binding and protein interactions .
C-terminal helical domain: Mediates telomere localization and interactions with telomeric proteins like TPP1 .
OBFC1 is a subunit of the CST complex (Ctc1-STN1-TEN1), which acts as a telomere-specific replication protein A (RPA) complex:
Telomere binding: Binds G-strand telomeric ssDNA with high affinity (K<sub>d</sub> ~50 nM) .
Telomere length regulation: Overexpression of OBFC1 mutants (e.g., ΔOB) causes telomere elongation in human cells .
Interaction with TPP1: Co-localizes with TPP1 via bimolecular fluorescence complementation (BiFC), requiring the C-terminal helical domain .
Genome-wide association studies (GWAS) link OBFC1 polymorphisms to leukocyte telomere length (LTL):
rs4387287: Strongest association (meta-analysis P = 2.3 × 10<sup>−11</sup>, N = 6,293) .
Ethnic variation: African populations exhibit longer telomeres, potentially due to OBFC1 haplotype differences .
DNA binding assays: Recombinant OBFC1 binds telomeric ssDNA (G-strand preference) in electrophoretic mobility shift assays (EMSAs) .
Cellular localization: Endogenous OBFC1 co-stains with RAP1 at telomeres in U2OS cells .
Functional disruption: Deletion of the OB fold reduces telomere localization but enhances TPP1 interaction .
Cancer: OBFC1 dysregulation is implicated in telomere elongation in cancer cells, promoting genomic instability .
Cardiovascular disease: A 2-SNP OBFC1 haplotype (rs10786775/rs11591710) associates with coronary heart disease (OR = 1.34, P = 0.04) .
Aging: GWAS data suggest OBFC1 variants account for ~1.2% of LTL variation in aging populations .
Telomere restriction fragment (TRF) assay: Used to quantify OBFC1-mediated telomere elongation in HTC75 cells .
Bimolecular fluorescence complementation (BiFC): Maps OBFC1-TPP1 interactions in live cells .
Recombinant OBFC1 production: E. coli-expressed His-tagged OBFC1 retains ssDNA-binding activity for in vitro studies .
OBFC1, also known as AAF44 (α-accessory factor 44), is a human homolog of yeast Stn1, a protein specifically involved in telomere replication and capping. OBFC1 contains an OB (oligonucleotide/oligosaccharide-binding) fold in its N-terminus and a helical domain in its C-terminus . The protein plays a crucial role in telomere maintenance by regulating telomere length and function in human cells. It achieves this through binding to telomeric single-stranded DNA and interacting with other telomere-associated proteins . Research has demonstrated that OBFC1 localizes to telomeres in human cells and participates in the complex network of protein interactions that protect and maintain chromosome ends .
OBFC1 consists of two main structural domains with distinct functions:
N-terminal OB fold domain: This domain shares significant homology with the yeast Stn1 protein and is responsible for binding to single-stranded DNA .
C-terminal helical domain: This region has no significant homology with other known motifs but is essential for stable telomere localization of OBFC1 .
The functional significance of this structure was demonstrated when deletion of the C-terminal helical domain prevented telomere localization of OBFC1, indicating that the OB fold alone is insufficient for stable telomere localization . This structural organization enables OBFC1 to coordinate both DNA binding and protein-protein interactions at telomeres.
Genome-wide association studies have identified several significant OBFC1 SNPs (single nucleotide polymorphisms) associated with leukocyte telomere length (LTL). Most notably:
rs4387287 showed significant association (P = 3.9 × 10⁻⁹) with LTL at a genome-wide significance level .
rs9419958 was also identified as significantly associated with telomere length .
Two nonsynonymous SNPs in tight linkage disequilibrium, rs2487999 (P = 2.7 × 10⁻⁸) and rs10786775 (P = 6.1 × 10⁻⁸, pairwise r² = 1.0), were found to have strong associations with telomere length .
These genetic variants demonstrate substantial effects on telomere length, with differences between genotypes averaging 230-400 bp depending on the specific variant and population studied . This magnitude of difference is considerable given that the average rate of LTL shortening in the general population is approximately 20-30 bp/year .
Research has demonstrated population and sex-specific differences in the effects of OBFC1 variants on telomere length. In one study involving multiple cohorts, the mean LTL difference across genotypes for rs437287 was 400 bp in men and 180 bp in women . In white populations, the difference across OBFC1 genotypes of rs4387287 and rs9419958 amounted to approximately 230 bp and 290 bp, respectively .
The following table summarizes some of the clinical characteristics of participants in studies examining OBFC1 variants:
| Parameter | Framingham Heart Study | Family Heart Study | Cardiovascular Health Study | Bogalusa Heart Study |
|---|---|---|---|---|
| Number | 1,146 | 877 | 1,061 | 333 |
| Women (%) | 51 | 51 | 62 | 42 |
| Age in years (range) | 59 (33–86) | 62 (30–93) | 75 (67–95) | 35 (20–48) |
| BMI (kg/m²) | 28.0 ± 5.0 | 29.1 ± 5.6 | 26.6 ± 4.4 | 28.0 ± 6.7 |
This variability suggests that the effects of OBFC1 variants on telomere dynamics may be modulated by sex-specific and potentially other demographic factors, highlighting the complexity of telomere regulation in humans .
To study OBFC1 localization to telomeres, researchers have employed several complementary approaches:
Indirect Immunofluorescence Analysis:
Bimolecular Fluorescence Complementation (BiFC) Assay:
Domain Analysis:
These methods provide complementary information about both the localization patterns and protein interaction networks of OBFC1 at telomeres.
To measure the impact of OBFC1 variants on telomere length, researchers should consider the following methodological approach:
Telomere Restriction Fragment (TRF) Assay:
Clone sequences encoding full-length and mutant human OBFC1 into appropriate vectors (e.g., pBabe-based retroviral vectors with FLAG tags)
Generate stable cell lines expressing these constructs through retroviral infection followed by selection
Allow cells to recover (designated as "P0") and passage for collection at various time points
Purify genomic DNA from cells using commercial kits (e.g., DNeasy kit)
Digest DNA with appropriate restriction enzymes (e.g., HinF1 and Rsa1)
Resolve the digested DNA by agarose gel electrophoresis
Transfer to nylon membranes for Southern blotting
Use radiolabeled telomeric probes (e.g., (TTAGGG)₃)
Analyze data using specialized software (e.g., Telorun analysis tool)
Genome-wide Association Study (GWAS) Approach:
Collect DNA samples from a large cohort (>3,000 individuals recommended)
Measure leukocyte telomere length (LTL) in all participants
Perform genome-wide genotyping
Identify SNPs associated with LTL variation
Attempt replication of top SNPs through de novo genotyping of additional individuals and in silico lookups in independent cohorts
These methodologies have been successfully used to identify and characterize the effects of OBFC1 variants on telomere length regulation.
OBFC1 interacts with multiple components of the telomere-maintenance machinery, particularly with TPP1, a core component of the telosome complex. This interaction was identified through large-scale immunoprecipitation and mass spectrometry analysis of TPP1 protein complexes in mammalian cells . The interaction between OBFC1 and TPP1 has been verified using bimolecular fluorescence complementation (BiFC) assays in live cells, which demonstrated that when OBFC1 and TPP1 interact, the N- and C-terminal YFP fragments come into close proximity, allowing fluorescence complementation to occur .
In addition to TPP1, OBFC1 likely interacts with other components of the telomere protein complex, including TRF1, TRF2, TIN2, RAP1, and POT1, which were all found in the TPP1 complex along with OBFC1 . The roles of OBFC1 in this complex network appear to mirror those of its yeast homolog Stn1, which regulates telomerase action on telomeres and coordinates DNA replication of the telomeric C strand .
From a mechanistic perspective, OBFC1:
Protects terminal telomeric DNA
Negatively regulates telomerase action on telomeres
Coordinates DNA replication of the opposing telomeric C strand
These interactions place OBFC1 as a central regulator in the complex network of telomere maintenance proteins.
OBFC1 has significant implications for aging-related diseases due to its role in telomere maintenance. Research has established that shortened leukocyte telomere length (LTL) is observed in various aging-related complex genetic diseases and is associated with diminished survival in elderly populations . The identification of OBFC1 as a locus for interindividual variation in LTL advances our understanding of telomere biology in humans and provides insights into aging-related disorders linked to altered telomere dynamics .
Several lines of evidence connect OBFC1 to aging-related diseases:
Rare mutations in genes that regulate telomere length have been identified in monogenic diseases such as dyskeratosis congenita and idiopathic pulmonary fibrosis, which are associated with shortened LTL and increased risk for aplastic anemia .
Shortened LTL denotes susceptibility to aging-related diseases, particularly atherosclerosis, in the general population .
The associations of LTL with OBFC1 and other telomere-related genes (e.g., TERC) support the hypothesis that telomere biology is an important pathway in human aging .
Therapeutic implications include:
Potential targeting of OBFC1 or its regulatory pathways to modulate telomere length in age-related diseases
Development of biomarkers based on OBFC1 variants to assess predisposition to accelerated telomere shortening
Personalized medicine approaches that consider OBFC1 genotypes in treatment strategies for age-related diseases
These findings collectively suggest that OBFC1 represents a promising target for understanding and potentially intervening in the aging process and age-related diseases.
When designing experiments to study the functional consequences of OBFC1 mutations, researchers should consider a comprehensive approach that includes:
Mutation Selection and Creation:
Focus on naturally occurring variants identified through GWAS (e.g., rs4387287, rs9419958, rs2487999, rs10786775)
Create targeted mutations in functional domains (particularly the OB fold and C-terminal helical domain)
Generate truncation mutations similar to those shown to cause telomere elongation in cancer cells
Cellular Models:
Functional Assays:
Telomere length measurement using TRF assays over multiple cell passages
Protein-protein interaction studies using co-immunoprecipitation, BiFC, or yeast two-hybrid approaches
DNA binding assays to assess interaction with telomeric single-stranded DNA
Telomerase activity assays to determine how OBFC1 variants affect telomerase function
Cell proliferation and senescence assays to assess biological consequences
In vivo Studies:
Generate mouse models with OBFC1 mutations corresponding to human variants
Assess telomere length and function across different tissues
Evaluate aging phenotypes and disease susceptibility
Data Analysis:
This multi-faceted experimental approach will provide comprehensive insights into how OBFC1 mutations affect telomere biology and cellular function.
When investigating OBFC1's role in telomere regulation, several critical controls and validation steps should be implemented:
Expression Controls:
Localization Validation:
Confirm telomere localization using multiple independent methods:
Interaction Verification:
Functional Redundancy Assessment:
Temporal Considerations:
Genetic Background Controls:
Technical Validation:
Use multiple telomere length measurement techniques (TRF, qPCR, Q-FISH)
Include internal controls for telomere length measurements
Perform technical replicates and ensure reproducibility across independent biological samples
When faced with contradictory findings regarding OBFC1's effects on telomere length, researchers should consider several factors in their interpretation:
Genetic Context Dependencies:
Methodological Variations:
Tissue and Cell Type Specificity:
Temporal Dynamics:
Protein Dosage Effects:
Mutation-Specific Effects:
Resolution Framework:
Design experiments that directly test competing hypotheses
Implement systematic replication studies in multiple independent laboratories
Consider meta-analysis approaches to integrate findings across multiple studies
By systematically evaluating these factors, researchers can develop a more nuanced understanding of OBFC1's complex role in telomere regulation and reconcile seemingly contradictory findings.
When analyzing OBFC1 genetic association data with telomere length, several statistical approaches should be considered to ensure robust and reliable findings:
Primary Association Analysis:
Multiple Testing Correction:
Replication Strategies:
Population Stratification Control:
Gene-Environment Interaction Analysis:
Longitudinal Analysis:
Apply mixed models for repeated telomere length measurements
Assess whether OBFC1 variants affect the rate of telomere shortening over time
Consider time-varying covariates in modeling longitudinal effects
Mediation Analysis:
Assess whether OBFC1 genetic effects on disease outcomes are mediated by telomere length
Implement formal statistical mediation tests
Calculate the proportion of genetic effect explained by telomere length variation
Power Calculations:
These statistical approaches have been successfully applied in previous studies of OBFC1 genetic associations and provide a robust framework for analyzing telomere genetics data.
Several emerging technologies hold promise for advancing our understanding of OBFC1's role in human telomere biology:
CRISPR-Cas9 Genome Editing:
Generate precise knockin models of OBFC1 variants identified in human populations
Create domain-specific mutations to dissect functional roles
Implement CRISPR interference (CRISPRi) or activation (CRISPRa) for temporal control of OBFC1 expression
Use base editing or prime editing for precise nucleotide substitutions without double-strand breaks
Single-Cell Telomere Analysis:
Apply single-cell sequencing technologies to understand cell-to-cell variation in telomere length
Investigate how OBFC1 variants affect telomere heterogeneity within tissues
Combine with single-cell transcriptomics to correlate OBFC1 expression with telomere dynamics
Advanced Imaging Techniques:
Implement super-resolution microscopy (STORM, PALM) to visualize OBFC1-telomere interactions
Use live-cell imaging with fluorescent telomere markers to track dynamics in real-time
Apply correlative light and electron microscopy (CLEM) to understand structural relationships
Proximity Labeling Methods:
Use BioID or APEX2 proximity labeling fused to OBFC1 to identify near-neighbor proteins
Map the complete telomeric interactome of OBFC1 under different conditions
Investigate changes in protein interactions caused by OBFC1 variants
Long-Read Sequencing:
Apply Oxford Nanopore or PacBio sequencing to characterize telomere structure and composition
Analyze telomere sequence variations in relation to OBFC1 genotypes
Develop new computational approaches for telomere assembly and analysis
Cryo-Electron Microscopy:
Determine high-resolution structures of OBFC1 in complex with telomeric DNA
Visualize conformational changes induced by different OBFC1 variants
Map the structural basis of OBFC1 interactions with other telomere proteins
Organoid and iPSC Models:
Generate induced pluripotent stem cells (iPSCs) from individuals with different OBFC1 genotypes
Differentiate into relevant tissue types to study tissue-specific effects
Create cerebral, cardiac, or other organoids to model telomere dynamics in complex tissues
These technologies, applied systematically to the study of OBFC1, will likely yield transformative insights into telomere biology and aging mechanisms in humans.
Research on OBFC1 could significantly contribute to developing interventions for telomere-related disorders through several potential translational pathways:
Precision Medicine Approaches:
Drug Discovery Targets:
Gene Therapy Strategies:
Develop gene editing approaches to correct pathogenic OBFC1 variants
Consider AAV-mediated delivery of functional OBFC1 in tissues affected by telomere dysfunction
Implement RNA therapies to modulate OBFC1 expression in specific tissues
Biomarker Development:
Cell-Based Therapies:
Generate stem cells with optimized OBFC1 function for transplantation
Engineer cells with enhanced telomere maintenance capacity for cellular therapy
Develop ex vivo expansion protocols that preserve telomere length in therapeutic cells
Lifestyle and Pharmaceutical Interventions:
Identify lifestyle modifications that may be particularly beneficial for individuals with specific OBFC1 variants
Develop pharmaceutical interventions that compensate for telomere dysfunction caused by OBFC1 variants
Test whether existing drugs (e.g., certain anti-inflammatory agents) have differential effects based on OBFC1 genotype
Combination Approaches:
The translational potential of OBFC1 research is significant given its central role in telomere biology and the association of telomere dysfunction with numerous age-related diseases including cardiovascular disease, pulmonary fibrosis, bone marrow failure, and certain cancers .
Despite significant advances in understanding OBFC1's function, several critical questions remain unresolved:
Resolving these questions will require integrative approaches combining structural biology, genetics, cell biology, and clinical research to fully understand OBFC1's complex role in human telomere maintenance and aging.
Oligonucleotide/oligosaccharide-binding (OB) fold is a structural motif found in a variety of proteins across different species, including prokaryotes and eukaryotes. This motif is known for its ability to bind single-stranded DNA (ssDNA) or RNA with high affinity. One of the notable proteins containing this motif is the human single-stranded DNA-binding protein 1 (hSSB1), which plays a crucial role in the DNA damage response (DDR).
The OB fold is characterized by a beta-barrel structure that forms a binding surface for nucleic acids. In hSSB1, the OB fold is located at the N-terminus and is responsible for binding to ssDNA. This binding is essential for the protein’s role in maintaining genomic stability, particularly during DNA replication and repair processes .
hSSB1 is rapidly recruited to sites of DNA damage, where it facilitates the formation of repair foci by interacting with other DDR proteins such as BRCA1 and RAD51. Recent studies have shown that the OB fold of hSSB1 also binds to poly(ADP-ribose) (PAR), a polymer synthesized by PARP enzymes in response to DNA damage . This interaction is crucial for the early recruitment of hSSB1 to DNA lesions, highlighting the importance of the OB fold in the DDR .
Given its critical role in DNA repair, hSSB1 and other OB fold-containing proteins are of significant interest in cancer research. Targeting these proteins could enhance the efficacy of existing therapies that induce DNA damage, such as radiation and certain chemotherapeutic agents . Additionally, understanding the mechanisms by which OB fold proteins contribute to genome stability could lead to the development of novel therapeutic strategies for diseases characterized by genomic instability .