The LBH gene spans ~28 kb, containing three exons that encode a 105-amino acid intrinsically disordered protein (IDP) . Key features include:
Protein characteristics: Acidic, nuclear-localized, and disordered, enabling dynamic interactions with diverse targets .
Conservation: Highly conserved across vertebrates, with no identified paralogs despite its multifunctionality .
| Feature | Detail |
|---|---|
| Gene length | 28,495 base pairs |
| Protein length | 105 amino acids |
| Molecular weight | 14.6 kDa (recombinant form) |
| Expression sites | Embryonic limb/heart, adult brain, kidney, spleen |
LBH acts as a transcriptional activator in signaling pathways, including Wnt/β-catenin and mitogen-activated protein kinase (MAPK) .
Wnt/β-catenin regulation: Downregulates Wnt signaling to modulate cell proliferation and differentiation .
MAPK signaling: Activates AP-1 and serum response element (SRE) transcription factors via ERK, JNK, and p38 pathways .
ΔNp63 induction: Promotes basal mammary stem cell differentiation and proliferation .
LBH is critical in embryogenesis and cancer progression, with context-dependent effects.
Limb/heart formation: Expressed in distal limb ectoderm and ventricular myocardium, overlapping with En1 and Fgf8 .
Cardiogenesis: Overexpression disrupts Nkx2.5 and Tbx5 expression, causing congenital heart defects .
LBH overexpression is linked to aggressive subtypes of multiple cancers:
Recent studies highlight LBH’s protective role in cardiac injury:
LBH-CRYAB signaling: Activates p38 phosphorylation and inhibits apoptosis/ferroptosis in cardiomyocytes during ischemia-reperfusion (I/R) injury .
Mechanistic Insight:
LBH’s dual roles in cancer and tissue protection present complex therapeutic challenges:
Oncology: Targeting LBH in basal breast cancer or glioma may inhibit tumor growth .
Cardioprotection: LBH upregulation could mitigate I/R injury, though risks of cancer progression must be weighed .
LBH was isolated from a human embryonic heart cDNA library. The LBH cDNA is 2,927 bp long, encoding a protein product of 105 amino acids. Research classifies LBH as an intrinsically disordered protein (IDP) that lacks a stable tertiary structure in isolation but undergoes a "disorder-to-order" transition upon binding to target molecules. This conformational flexibility allows LBH to potentially acquire different functional activities depending on the specific target-induced changes, which is characteristic of many transcriptional regulators . Experimental approaches to study LBH structure typically include circular dichroism spectroscopy, nuclear magnetic resonance, and in silico molecular modeling to capture its conformational dynamics.
LBH expression regulation involves complex mechanisms centered around the Wnt signaling pathway. To investigate this regulation, researchers should implement a methodological approach including: (1) Promoter analysis using reporter assays to identify regulatory elements; (2) Chromatin immunoprecipitation (ChIP) to detect transcription factor binding; (3) DNA methylation analysis to assess epigenetic regulation; and (4) RNA stability assays to evaluate post-transcriptional regulation. When examining tissue-specific expression patterns, researchers should employ both qRT-PCR and immunohistochemistry across multiple tissue types, with careful attention to developmental stage variability .
LBH functions as an important transcriptional regulator in the wingless/int-1 (Wnt) signaling pathway. Research indicates that LBH can suppress mammary epithelial cell differentiation and potentially contribute to Wnt-induced tumorigenesis . To properly investigate LBH functions, researchers should implement gain-of-function and loss-of-function studies using techniques such as CRISPR-Cas9 genome editing, siRNA knockdown, and overexpression systems. These approaches should be coupled with phenotypic assays measuring proliferation, differentiation, migration, and apoptosis to comprehensively characterize LBH's functional impact in specific cellular contexts.
The interaction between LBH and the Wnt signaling pathway represents a complex relationship with significant implications for development and disease. Methodologically, researchers should approach this question through multiple complementary techniques: (1) Co-immunoprecipitation and proximity ligation assays to identify direct protein-protein interactions; (2) ChIP-seq to map LBH binding sites on chromatin in relation to Wnt-responsive elements; (3) RNA-seq following LBH modulation to identify downstream transcriptional effects; and (4) Reporter assays using TCF/LEF binding elements to measure Wnt pathway activity. In cancer contexts, particular attention should be paid to β-catenin localization and activity, as this represents a critical node in canonical Wnt signaling that may be influenced by LBH .
While the search results don't explicitly detail post-translational modifications (PTMs) of LBH, this represents an important research direction given LBH's role as an intrinsically disordered protein. To investigate PTMs of LBH, researchers should employ: (1) Mass spectrometry-based proteomics to identify specific modification sites; (2) Site-directed mutagenesis to create modified variants; (3) Phospho-specific antibodies to track dynamic modifications; and (4) Cellular fractionation studies to assess how modifications affect subcellular localization. The experimental design should include multiple cell types and conditions (normal vs. stress vs. disease states) to capture context-dependent regulation of LBH function through PTMs.
To comprehensively map LBH's protein interaction network, researchers should implement multi-layered approaches: (1) Affinity purification coupled with mass spectrometry (AP-MS) to identify stable interactors; (2) BioID or APEX proximity labeling to capture transient interactions; (3) Yeast two-hybrid screening for direct binary interactions; and (4) Co-immunoprecipitation with candidate partners identified through bioinformatic prediction. These protein-protein interaction studies should be conducted in relevant cellular contexts, particularly those where LBH demonstrates functional significance, such as hepatocellular carcinoma tissues or breast cancer cell lines . Network analysis tools should then be applied to integrate these findings with known signaling pathways.
When designing experiments to investigate LBH expression in human cancers, researchers must consider several methodological aspects:
Sample selection: Include adequate numbers of tumor tissues with matched normal adjacent tissues (minimum n=100 for statistical power)
Patient cohort: Ensure diverse representation across clinical stages, demographic factors, and treatment histories
Expression analysis: Implement multiple detection methods including:
Immunohistochemistry with validated antibodies and appropriate controls
qRT-PCR with reference gene normalization
Western blotting for protein quantification
Scoring system: Establish consistent scoring criteria for expression levels (e.g., H-score or percentage of positive cells)
Clinical correlation: Collect comprehensive clinical data including disease stage, biomarkers, and patient outcomes
This approach mirrors the methodology used in HCC research where 226 patient samples were analyzed for LBH expression with correlation to clinical parameters and survival outcomes .
When using cell line models to study LBH function, researchers must implement rigorous controls and validation:
Expression verification: Confirm baseline LBH expression across multiple cell lines using qRT-PCR and Western blotting
Genetic manipulation controls:
For overexpression: Empty vector controls processed identically
For knockdown: Non-targeting siRNA/shRNA controls
For CRISPR: Non-targeting gRNA controls
Rescue experiments: Re-express wild-type LBH in knockout models to confirm phenotype specificity
Multiple cell line validation: Replicate key findings in at least 3 different cell lines
Authentication: Regular STR profiling and mycoplasma testing
Physiological relevance: Compare expression levels to those observed in primary human tissues
Functional assays: Include positive and negative controls specific to each assay (proliferation, migration, etc.)
For reliable quantification of LBH expression, researchers should consider multiple complementary techniques:
| Technique | Advantages | Limitations | Best Application |
|---|---|---|---|
| Immunohistochemistry (IHC) | Preserves tissue architecture; allows cellular localization | Semi-quantitative; dependent on antibody quality | Tissue expression patterns and localization |
| Western blotting | Detects specific protein forms; semi-quantitative | Requires tissue lysis; loses spatial information | Protein expression comparison between samples |
| qRT-PCR | Highly sensitive; good for low abundance detection | Measures mRNA not protein; potential primer bias | mRNA expression screening across many samples |
| RNA-seq | Global transcriptomic context; isoform detection | Cost; complex analysis; mRNA not protein | Transcriptional profiling and isoform analysis |
| Proteomics (MS) | Unbiased; can detect modifications | Complex sample preparation; expensive | Protein modifications and interaction studies |
The study design should incorporate at least two orthogonal methods for validation, as seen in the HCC research where IHC findings were correlated with clinical biochemical markers .
When confronted with conflicting data about LBH expression across cancer types, researchers should implement a systematic analytical approach:
Context evaluation: Assess biological context differences (tissue type, disease stage, molecular subtypes)
Methodological comparison: Evaluate differences in detection methods, antibodies, and quantification approaches
Statistical reassessment: Consider sample sizes, power calculations, and potential confounding variables
Meta-analysis: Perform quantitative synthesis of published data with subgroup analysis
Heterogeneity assessment: Investigate tumor heterogeneity through single-cell approaches or microdissection
Technical validation: Replicate key findings using orthogonal techniques
Functional correlation: Relate expression differences to functional outcomes in cellular models
This methodological framework helps distinguish biological variation from technical artifacts, providing clearer interpretation of apparently conflicting results across cancer types.
Based on research methodologies in LBH cancer studies, the following statistical approaches are recommended:
Survival analysis:
Expression thresholds:
ROC curve analysis to determine clinically relevant cutoff values
Quantile-based grouping (tertiles/quartiles) to examine dose-response relationships
Continuous analysis to avoid information loss from dichotomization
Advanced approaches:
Propensity score matching to reduce selection bias
Landmark analysis to address time-dependent factors
Machine learning algorithms for complex pattern recognition
Key considerations should include adequate sample size (minimum 200 patients for survival analysis), appropriate follow-up duration (minimum 5 years for cancer outcomes), and validation in independent cohorts.
Differentiating correlation from causation in LBH research requires methodological rigor:
Temporal sequence establishment:
Longitudinal studies with multiple timepoints
Early vs. late stage comparison within same cancer type
Pre-malignant to malignant progression models
Dose-response relationship:
Graded expression models (knockdown, wild-type, overexpression)
Titration experiments with inducible systems
Correlation of expression levels with phenotype intensity
Mechanistic validation:
Pathway perturbation experiments
Rescue experiments reversing phenotypic effects
Identification of direct molecular targets
Causal inference methods:
Mendelian randomization using genetic instruments
Mediation analysis to identify intermediate variables
Directed acyclic graphs (DAGs) to model causal relationships
Animal models:
Genetically engineered models with tissue-specific LBH modulation
Xenograft studies with LBH-modified cells
Pharmacological intervention targeting LBH-dependent pathways
LBH overexpression demonstrates significant prognostic value in hepatocellular carcinoma (HCC). Research findings indicate that high levels of LBH could be detected in 8.8% (20/226) of HCC samples. Correlation analysis demonstrated that LBH protein levels in HCC were significantly associated with serum AST/ALT levels and clinical stage, suggesting a relationship with liver dysfunction and disease progression. Most importantly, Kaplan-Meier survival analysis revealed that patients with low LBH expression had significantly longer mean survival times compared to those with high LBH expression .
These findings suggest LBH may serve as an independent prognostic biomarker in HCC. For clinical implementation, researchers should further validate these findings through:
Multivariate analysis controlling for established prognostic factors
Standardization of LBH assessment methods
Prospective validation in independent patient cohorts
Correlation with treatment response parameters
Developing LBH as a therapeutic target requires a multifaceted research approach:
Target validation:
Demonstrate addiction to LBH in relevant cancer models
Identify synthetic lethal interactions with LBH expression
Establish reversibility of malignant phenotypes upon LBH inhibition
Intervention strategies:
Small molecule inhibitors targeting LBH-protein interactions
Peptide mimetics disrupting key functional domains
RNA-based therapeutics (siRNA, antisense oligonucleotides)
Proteolysis-targeting chimeras (PROTACs) for LBH degradation
Precision medicine considerations:
Identify patient subgroups most likely to benefit from LBH targeting
Develop companion diagnostics for LBH expression/activity
Investigate combination strategies with standard treatments
Predictive biomarkers:
Establish LBH expression thresholds for treatment response
Identify downstream markers of effective LBH targeting
Develop pharmacodynamic markers for dose optimization
Given LBH's role in the Wnt pathway and its association with poor prognosis in HCC, targeting the LBH-Wnt axis represents a promising therapeutic strategy worth further investigation .
Translating LBH research to clinical applications faces several methodological and practical challenges:
Biological complexity:
Context-dependent functions across different tissues
Redundancy in signaling pathways
Potential for adaptive resistance mechanisms
Technical limitations:
Specificity of detection methods across diverse sample types
Standardization of expression assessment between laboratories
Development of clinically validated antibodies or assays
Clinical validation barriers:
Need for large, prospective clinical trials
Patient stratification criteria
Integration with existing prognostic models
Therapeutic development challenges:
Targeting transcription factors effectively
Achieving sufficient specificity to minimize off-target effects
Developing appropriate drug delivery systems
Regulatory considerations:
Biomarker validation requirements
Companion diagnostic development
Clinical trial design for targeted therapies
Researchers must address these challenges through collaborative efforts between basic scientists, translational researchers, and clinicians to bridge the bench-to-bedside gap.
To push LBH research boundaries, investigators should consider implementing:
Single-cell technologies:
scRNA-seq to profile LBH expression heterogeneity
Single-cell ATAC-seq to map chromatin accessibility
Single-cell proteomics for protein-level analysis
Spatial transcriptomics to preserve tissue context
Advanced protein analysis:
Hydrogen-deuterium exchange mass spectrometry for structural dynamics
Cross-linking mass spectrometry for interaction interfaces
AlphaFold2 and other AI-based structural prediction tools
Protein painting for mapping functional domains
Functional genomics:
CRISPR screens (knockout, activation, inhibition) to identify synthetic interactions
Base editing for precise genetic modifications
CRISPR-based lineage tracing in development and disease progression
Pooled CRISPR screens with single-cell readouts
Translational technologies:
Patient-derived organoids to model LBH function in personalized models
Microfluidic devices for high-throughput drug screening
In situ sequencing for spatial mapping of LBH and interacting partners
Liquid biopsy approaches to track LBH-related biomarkers
Investigating LBH's role in metastasis requires a comprehensive experimental design:
Clinical correlation:
Compare LBH expression between primary tumors and matched metastases
Analyze correlation between LBH levels and metastatic burden
Perform subgroup analysis based on metastatic site
In vitro functional assays:
Invasion assays (transwell, 3D matrix invasion)
Migration assays (wound healing, single-cell tracking)
Adhesion assays to different extracellular matrices
Epithelial-mesenchymal transition marker analysis
Ex vivo approaches:
Circulating tumor cell isolation and characterization
Patient-derived explant cultures with metastatic capacity
In vivo metastasis models:
Orthotopic implantation with spontaneous metastasis
Experimental metastasis via tail vein or intracardiac injection
Fluorescent/bioluminescent imaging for tracking
Analysis of pre-metastatic niche formation
Molecular mechanism investigations:
RNA-seq to identify metastasis-specific transcriptional programs
ChIP-seq to map LBH binding sites in metastatic vs. non-metastatic cells
Proteomics to identify metastasis-specific interaction partners
CRISPR screens for synthetic lethal interactions in metastatic cells
Multi-omic integration:
Integrate transcriptomic, epigenomic, and proteomic data
Network analysis to identify critical nodes in metastatic processes
Validate key findings across multiple experimental models and clinical samples
Studying LBH's conformational dynamics as an intrinsically disordered protein requires specialized controls:
Protein preparation controls:
Multiple purification methods to ensure native structure preservation
Size exclusion chromatography to confirm monomeric state
Circular dichroism to verify disordered characteristics
Tag position variations to minimize interference
Binding partner controls:
Mutated binding interfaces to demonstrate specificity
Concentration gradients to establish binding kinetics
Competition assays with known partners
Non-binding protein controls of similar size/charge
Structural analysis controls:
Temperature and pH series to assess stability
Chemical denaturants to establish folding/unfolding baselines
Paramagnetic relaxation enhancement controls in NMR studies
Crosslinking distance controls for validation
Functional correlation controls:
Structure-guided mutations affecting transition without disrupting expression
Domain swap experiments to identify critical regions
Correlation of structural changes with functional readouts
Time-resolved studies to capture transition kinetics
Computational controls:
Multiple force fields in molecular dynamics simulations
Ensemble approaches rather than single-structure models
Validation across different simulation time scales
Comparison with experimentally determined parameters
Based on current knowledge gaps, researchers should prioritize these emerging questions:
Mechanistic understanding:
What is the complete interactome of LBH in normal vs. disease states?
How does LBH specifically regulate transcription at the molecular level?
What are the structural determinants of LBH's disorder-to-order transitions?
How is LBH activity regulated post-translationally?
Disease relevance:
Beyond HCC and breast cancer, what other cancer types show LBH dysregulation?
Does LBH play roles in non-cancer pathologies?
Can LBH serve as a pan-cancer prognostic biomarker?
Are there LBH polymorphisms associated with disease susceptibility?
Therapeutic potential:
What are the druggable nodes in LBH-dependent pathways?
Can LBH status predict response to existing therapies?
Would targeting LBH synergize with current standard-of-care treatments?
How can LBH-based biomarkers be implemented in clinical decision-making?
Developmental biology:
What is LBH's role in embryonic development beyond heart and limb formation?
How does LBH function in adult tissue homeostasis?
Does LBH contribute to stem cell maintenance or differentiation?
What evolutionary conservation exists in LBH function across species?
Computational approaches offer powerful tools for advancing LBH research:
Structural prediction and analysis:
AI-based structure prediction (AlphaFold2, RoseTTAFold)
Molecular dynamics simulations of disorder-to-order transitions
Virtual screening for potential binding partners or inhibitors
Modeling of conformational ensembles rather than static structures
Network biology:
Pathway enrichment and interaction network analysis
Master regulator analysis to position LBH in regulatory hierarchies
Causal network inference from multi-omic data
Network perturbation models to predict therapeutic responses
Translational bioinformatics:
Multi-cancer expression analysis across public datasets (TCGA, GEO)
Survival prediction models incorporating LBH expression
Drug response prediction based on LBH-associated signatures
Patient stratification algorithms for clinical applications
AI and machine learning:
Deep learning for image analysis of LBH staining patterns
Natural language processing to extract LBH knowledge from literature
Reinforcement learning for optimal experimental design
Multi-modal data integration across experimental platforms
Systems biology:
Genome-scale metabolic modeling to connect LBH to metabolic changes
Multi-scale modeling linking molecular events to tissue-level phenomena
Feedback loop identification and analysis
Simulation of treatment effects on LBH-dependent systems
Current evidence establishes LBH as an important transcriptional regulator with significant implications in cancer biology. LBH plays a crucial role in the Wnt signaling pathway and can suppress mammary epithelial cell differentiation, potentially contributing to tumorigenesis. In hepatocellular carcinoma, high LBH expression correlates with poorer clinical outcomes, including shorter survival times . Additionally, LBH has been highlighted as a potential marker for therapeutically challenging basal-like breast cancer.
The intrinsically disordered nature of LBH protein suggests a complex and context-dependent function, likely involving disorder-to-order transitions upon binding to different molecular partners. This structural flexibility may underlie LBH's diverse roles across different tissues and disease states.
Despite these advances, significant knowledge gaps remain regarding LBH's precise molecular mechanisms, comprehensive interactome, and roles in cancer types beyond HCC and breast cancer. The field is advancing rapidly, with new techniques offering opportunities to deepen our understanding of this important regulator.
To advance LBH research effectively, investigators should adhere to these methodological recommendations:
Standardization practices:
Establish validated antibodies and detection protocols
Define consistent scoring systems for expression analysis
Create reference standards for quantitative comparisons
Develop reproducible experimental models
Comprehensive approach:
Implement multi-omic profiling (genomics, transcriptomics, proteomics)
Utilize both in vitro and in vivo models with clinical validation
Integrate computational and experimental methodologies
Study LBH in both physiological and pathological contexts
Collaborative frameworks:
Establish multi-institutional biobanking for diverse sample access
Create data sharing platforms for LBH-related findings
Develop interdisciplinary teams spanning basic science to clinical research
Engage patient advocates for outcome measure relevance
Technological advancement:
Apply cutting-edge single-cell and spatial biology techniques
Develop improved tools for studying intrinsically disordered proteins
Create better models recapitulating the tumor microenvironment
Implement AI/ML approaches for data integration and hypothesis generation
LBH Human Recombinant is produced in Escherichia coli (E. coli) and is a single, non-glycosylated polypeptide chain. It contains 128 amino acids and has a molecular mass of approximately 14.6 kDa . The recombinant protein is fused to a 23 amino acid His-tag at the N-terminus, which aids in its purification through chromatographic techniques .
The LBH protein is essential for the proper formation of limb buds and the heart during the early stages of human development. It is involved in the regulation of gene expression and cellular differentiation, which are critical processes for the development of these organs. Mutations or dysregulation of the LBH gene can lead to developmental abnormalities and congenital defects.
The recombinant LBH protein is produced using E. coli expression systems. The protein is then purified to a high degree of purity, typically greater than 90%, using proprietary chromatographic techniques . The purified protein is formulated as a sterile filtered clear solution containing 20 mM Tris-HCl (pH 8.0), 20% glycerol, 0.15 M NaCl, and 1 mM DTT .
LBH Human Recombinant protein is used in various research applications, including: