TXNL4A (thioredoxin-like 4A) is a human gene located on chromosome 18q23 that encodes a 19.3 kDa protein critical for pre-mRNA splicing. It is a core component of the U5 small nuclear ribonucleoprotein (snRNP) complex, which is essential for spliceosome assembly and catalytic activation . Mutations in TXNL4A are associated with Burn-McKeown syndrome (BMKS), a congenital disorder characterized by craniofacial abnormalities, hearing loss, and heart defects .
The TXNL4A protein facilitates RNA-RNA interactions within the major spliceosome, enabling precise removal of introns from pre-mRNA. Key functions include:
Spliceosome Assembly: Mediates structural rearrangements in U4/U6-U5 tri-snRNP complexes .
Catalytic Activation: Coordinates splice site recognition and exon ligation .
Compound heterozygosity is the primary genetic mechanism:
Loss-of-Function Mutations: Nonsense, frameshift, or microdeletions in one allele .
Promoter Deletions: 34 bp deletions in the core promoter region reduce protein expression .
Genetic Finding | Clinical Correlation | Reference |
---|---|---|
Compound heterozygosity | Craniofacial dysmorphisms, hearing loss | |
Homozygous promoter deletion | Severe BMKS in consanguineous families |
Recent studies identified TXNL4A as a prognostic biomarker in HCC:
Clinical Parameter | TXNL4A High Expression | Significance |
---|---|---|
T-stage | p < 0.001 | Advanced disease |
Vascular Invasion | p = 0.022 | Metastatic risk |
CD8 T Cell Infiltration | Positive correlation | Immune modulation |
The LOVD database reports 38 public variants, including:
Functional Impact: Reduced TXNL4A disrupts spliceosome assembly, affecting craniofacial development .
TXNL4A encodes a critical subunit of the major spliceosome, specifically functioning as a component of the U5 snRNP and U4/U6-U5 tri-snRNP complexes. The protein plays an essential role in spliceosome assembly and participates in the precatalytic spliceosome. Its primary function involves pre-mRNA processing, where it helps recognize and remove intronic regions to produce mature mRNA molecules. The TXNL4A protein is thought to prevent premature spliceosome activation, with its departure defining the transition from the B complex to the B act complex during the splicing cycle . The gene shows remarkable evolutionary conservation, with its yeast ortholog DIB1 encoding a small protein essential for pre-mRNA splicing, suggesting its fundamental importance in eukaryotic molecular biology .
When TXNL4A expression is reduced, research indicates defective assembly of the U4/U6.U5 tri-snRNP complex, which consequently disrupts normal splicing patterns. Interestingly, not all transcripts are equally affected by TXNL4A deficiency. Studies in S. cerevisiae with reduced DIB1 (the TXNL4A ortholog) expression revealed specific subsets of genes that were mis-expressed and/or mis-spliced compared to wildtype yeast, with the affected introns sharing conserved physical properties . This suggests TXNL4A deficiency creates selective vulnerability in certain RNA processing pathways rather than causing global splicing disruption. In human disease contexts, reduced TXNL4A expression is thought to specifically affect the splicing of transcripts crucial for craniofacial development, explaining the tissue-specific manifestations despite the gene's ubiquitous expression .
TXNL4A stands out among spliceosomal components due to its role as a regulatory element that governs spliceosome activation timing. Unlike many core spliceosomal proteins that directly catalyze splicing reactions, TXNL4A functions as a checkpoint protein that prevents premature spliceosome activation. This timing function is critical, as premature activation could lead to inefficient or incorrect splicing. The protein demonstrates high evolutionary conservation across species, with orthologs in yeast (DIB1), nematodes, and mammals all showing essential roles in splicing . This extreme conservation suggests TXNL4A performs a fundamental function that has remained largely unchanged throughout eukaryotic evolution. Additionally, its involvement in developmental disorders despite being part of the core splicing machinery highlights how disruption of basic cellular processes can manifest in tissue-specific ways during development.
Burn-McKeown syndrome (BMKS) arises from compound heterozygous mutations in the TXNL4A gene. Most affected individuals have different genetic changes in each copy of the TXNL4A gene: one copy typically contains a mutation that impairs protein function or prevents protein production entirely, while the other copy has a deletion in the promoter region that reduces protein expression . The combined effect of these mutations is a significant reduction in functional TXNL4A protein, which impairs spliceosome assembly and changes the production of specific mRNA molecules. Given the high homology between DIB1 (yeast ortholog) and TXNL4A, research suggests that the reduced TXNL4A expression affects assembly of the human tri-snRNP complex, which in turn disrupts splicing of pre-mRNAs critical for craniofacial development . This selective effect on particular developmental pathways explains why BMKS manifests primarily as craniofacial malformations despite TXNL4A's ubiquitous expression and fundamental cellular role.
TXNL4A mutations are associated with Burn-McKeown syndrome, a congenital disorder characterized by a specific constellation of clinical features. The primary manifestations include abnormalities of the nasal passages, distinctive facial features, hearing loss, cardiac abnormalities, and short stature . The craniofacial features are particularly distinctive, suggesting that craniofacial development is especially sensitive to spliceosome dysfunction. This sensitivity appears to be a common theme, as mutations in several genes involved in the spliceosome have been shown to cause conditions with craniofacial malformations . Research models using induced pluripotent stem cells have demonstrated that TXNL4A variants specifically influence neural crest cell function and behavior, providing a mechanistic link between the molecular defect and the tissue-specific manifestations . The specificity of these clinical features, despite TXNL4A's fundamental role in all cells, represents an important example of how disruption of basic cellular processes can result in tissue-specific developmental abnormalities.
For analyzing TXNL4A expression in clinical samples, researchers employ multiple complementary techniques for comprehensive assessment. RNA sequencing (RNA-Seq) provides quantitative measurement of TXNL4A transcript levels and allows detection of aberrant splicing events resulting from TXNL4A dysfunction. For validation of differential expression between normal and diseased tissues, quantitative PCR (qPCR) offers targeted verification with high sensitivity . Several large-scale databases can be utilized for expression analysis across different tissues and disease states, including TCGA-LIHC (The Cancer Genome Atlas-Liver Hepatocellular Carcinoma), ICGC (International Cancer Genome Consortium), GEPIA (Gene Expression Profiling Interactive Analysis), and UALCAN databases . For protein-level analysis, immunohistochemistry provides spatial information about TXNL4A expression within tissue architecture. In cancer research specifically, researchers have successfully used LASSO regression analysis to construct prognostic models incorporating TXNL4A expression . Additionally, Chi-squared analyses and Fisher tests can determine relationships between TXNL4A expression and clinical characteristics such as TNM stage, pathologic stage, and histologic grade .
Single-cell RNA sequencing (scRNA-seq) provides unique insights into TXNL4A function by revealing cell type-specific expression patterns and effects that would be masked in bulk tissue analysis. This approach has proven particularly valuable for investigating TXNL4A's role in immune cell populations within the tumor microenvironment. Recent studies analyzing scRNA-seq datasets (GSE140228_10X and GSE166635) demonstrated that TXNL4A is differentially expressed across immune cell populations in hepatocellular carcinoma, with particularly high expression in B cells, CD4 T cells, CD8 T cells, and monocytes/macrophages . These cell type-specific expression patterns provide critical context for understanding TXNL4A's influence on immune infiltration in cancer. Additionally, scRNA-seq can detect subtle alterations in gene expression and splicing patterns across developmental trajectories, offering insights into how TXNL4A mutations affect specific cell populations during embryonic development. For researchers investigating TXNL4A function, scRNA-seq databases such as TISCH2 provide valuable resources to analyze expression patterns across diverse cell types and disease states .
Developing appropriate experimental models for TXNL4A-related disorders requires careful consideration of the gene's essential nature and tissue-specific effects. Since complete knockout of TXNL4A orthologs in model organisms (S. cerevisiae, S. pombe, and C. elegans) is lethal, researchers must employ more nuanced approaches . Induced pluripotent stem cells (iPSCs) derived from patients with TXNL4A mutations provide an excellent model system, allowing investigation of how TXNL4A variants affect neural crest cell function and behavior, which is critical for understanding craniofacial development abnormalities . These iPSC models can be differentiated into neural crest cells and other relevant lineages to study developmental trajectories affected by TXNL4A dysfunction. For genetic manipulation, CRISPR/Cas9 systems with inducible or partial knockdown of TXNL4A can mimic the reduced expression seen in patient conditions while avoiding lethality. Conditional knockout models in mice, where TXNL4A expression is eliminated in specific tissues or at particular developmental stages, could provide insights into tissue-specific requirements for TXNL4A. Additionally, yeast models with reduced DIB1 expression have proven useful for studying fundamental mechanisms of how diminished TXNL4A function affects splicing of specific subsets of transcripts .
TXNL4A expression demonstrates significant correlations with immune cell infiltration in hepatocellular carcinoma, suggesting an immunomodulatory role. Multiple independent analytical approaches have confirmed this relationship. Analysis using the TIMER database revealed that TXNL4A expression positively correlates with infiltration of B cells, CD4 T cells, CD8 T cells, macrophages, neutrophils, and dendritic cells in HCC . These findings were further validated using the CIBERSORT algorithm, which showed higher levels of CD8 T cells, regulatory T cells, and M0 macrophages in tumors with high TXNL4A expression . The Gene Set Cancer Analysis (GSCA) database confirmed these patterns, showing positive correlations between TXNL4A expression and the infiltration of B cells, CD8 T cells, and dendritic cells . Single-cell RNA sequencing analyses provided additional resolution, demonstrating that TXNL4A is highly expressed in specific immune populations, particularly CD8 T cells, B cells, CD4 T cells, and monocytes/macrophages . The mechanistic basis for these correlations remains under investigation, but evidence suggests that abnormal RNA splicing, influenced by TXNL4A, may modulate immune responses by altering cytokine signaling efficiency . These findings highlight the potential significance of TXNL4A in cancer immunobiology and suggest possibilities for therapeutic targeting.
The tissue-specific manifestations of TXNL4A deficiency, particularly in craniofacial development, represent a fascinating paradox given the gene's ubiquitous expression and fundamental cellular role. Several converging mechanisms likely explain this specificity. First, RNA sequencing analysis has revealed that not all introns are equally affected by reduced TXNL4A function; instead, mis-spliced introns share specific physical properties that make them particularly vulnerable to spliceosomal defects . Transcripts containing these vulnerable introns may be enriched in developing craniofacial tissues. Second, neural crest cells, which contribute extensively to craniofacial structures, appear particularly sensitive to TXNL4A dysfunction, as demonstrated by studies using iPSC models . This sensitivity may stem from the demanding gene expression programs these cells must execute during migration and differentiation. Third, developmental timing creates windows of vulnerability during which even subtle defects in RNA processing can have profound effects on morphogenesis. The craniofacial structures develop during specific embryonic periods when precise gene expression is critical, making them vulnerable to splicing perturbations. Finally, this pattern of tissue-specific manifestations despite ubiquitous expression is observed with mutations in several spliceosomal genes, suggesting a common vulnerability in craniofacial development to spliceosomal dysfunction .
The emerging understanding of TXNL4A's role in both developmental disorders and cancer suggests several potential therapeutic avenues. For hepatocellular carcinoma, where TXNL4A appears to function as an oncogene, inhibiting its expression or activity could offer therapeutic benefits. Since TXNL4A is associated with docetaxel resistance pathways in HCC, combining TXNL4A inhibition with docetaxel treatment might enhance chemotherapeutic efficacy . TXNL4A's position in the RNA splicing pathway also makes it a promising target for RNA shear-targeting drugs or RNA vaccines, which represent cutting-edge approaches in cancer therapy . For developmental disorders like Burn-McKeown syndrome, therapeutic approaches would need to enhance TXNL4A function or compensate for its deficiency. This might involve targeted delivery of functional TXNL4A to affected tissues during critical developmental windows, potentially using gene therapy approaches. Alternatively, modulating downstream pathways affected by TXNL4A deficiency could mitigate developmental abnormalities. Given TXNL4A's role in regulating spliceosome assembly and activation, small molecules that stabilize the spliceosome or promote correct splicing of vulnerable transcripts might counteract the effects of TXNL4A mutations. As research advances, patient-derived iPSC models will likely prove invaluable for screening potential therapeutic compounds before clinical application .
Identifying TXNL4A-affected splice variants requires sophisticated bioinformatic approaches that can detect subtle changes in splicing patterns. Differential splicing analysis tools such as rMATS, SUPPA2, or LeafCutter can identify alternative splicing events (exon skipping, intron retention, alternative 5' or 3' splice sites) that differ between normal and TXNL4A-deficient samples. For comprehensive analysis of RNA-Seq data from TXNL4A-deficient models, researchers should implement splice junction-aware aligners like STAR or HISAT2, followed by transcript assembly tools such as StringTie or Cufflinks . Motif analysis of mis-spliced introns has proven valuable in understanding splice site vulnerability to TXNL4A deficiency, as studies in yeast revealed that mis-spliced introns share conserved physical properties . Researchers should also employ visualization tools like IGV or UCSC Genome Browser to manually inspect splicing anomalies at specific loci of interest. For identifying common features of TXNL4A-sensitive transcripts, enrichment analysis of Gene Ontology terms or pathways among mis-spliced genes can reveal biological processes particularly affected by TXNL4A deficiency. In cancer research contexts, GSEA (Gene Set Enrichment Analysis) has successfully identified pathways enriched in high-TXNL4A expression samples, including docetaxel resistance, embryonic stem cells, and tumor invasion pathways .
Integrating multi-omics data provides a comprehensive understanding of TXNL4A functions across molecular scales. Researchers should combine transcriptomics (RNA-Seq) with proteomics to correlate changes in splice variants with altered protein isoform expression, providing insights into functional consequences of TXNL4A-mediated splicing alterations. Chromatin immunoprecipitation sequencing (ChIP-seq) can identify transcriptional regulators of TXNL4A expression, helping to understand its dysregulation in cancer. For mechanistic insights, protein-protein interaction data from techniques like IP-MS (immunoprecipitation-mass spectrometry) can map TXNL4A's interactions within the spliceosome and beyond. In cancer research, integrating TXNL4A expression data with immunological profiles using techniques like CIBERSORT, TIMER, and single-cell RNA sequencing provides multidimensional understanding of TXNL4A's role in immune infiltration . For clinical applications, combining TXNL4A expression data with patient metadata, treatment responses, and survival outcomes enables development of comprehensive predictive models. Software platforms like MultiOmics Factor Analysis (MOFA) or Similarity Network Fusion (SNF) can integrate these diverse data types to identify patterns not apparent in single-omics analyses. When analyzing developmental disorders, researchers should integrate transcriptomics with imaging data from model organisms to correlate molecular changes with morphological outcomes. This multi-omics integration is particularly valuable for understanding how TXNL4A influences both cancer progression and developmental processes.
Developing experimental therapeutics targeting TXNL4A requires careful consideration of its dual roles in development and cancer. For cancer applications, where TXNL4A appears to function as an oncogene, inhibitory approaches are appropriate. These might include antisense oligonucleotides or siRNAs targeting TXNL4A mRNA, small molecules disrupting TXNL4A protein interactions within the spliceosome, or PROTAC (proteolysis targeting chimera) approaches to induce TXNL4A degradation. Given TXNL4A's role in RNA splicing, modulating its activity rather than completely eliminating it would be prudent to minimize off-target effects on essential cellular processes . For developmental disorders caused by TXNL4A deficiency, therapeutic approaches would aim to restore function. This might involve antisense oligonucleotides that promote correct splicing of affected transcripts, small molecules that stabilize spliceosome components to compensate for reduced TXNL4A, or gene therapy approaches delivering functional TXNL4A during critical developmental windows. For any therapeutic approach, researchers must consider potential tissue-specific effects and develop delivery methods that target relevant tissues: liver for HCC applications or neural crest cells for developmental disorders . Thorough preclinical testing should include assessment of splicing changes across the transcriptome to identify potential off-target effects. Finally, researchers should explore combination strategies, particularly in cancer contexts, where TXNL4A inhibition might sensitize tumors to existing therapeutics by modulating docetaxel resistance or immune infiltration pathways .
Optimizing animal models for human TXNL4A-related pathologies requires sophisticated genetic approaches that recapitulate the specific molecular defects seen in patients. For Burn-McKeown syndrome, compound heterozygous models combining a loss-of-function mutation with a promoter deletion would most accurately reflect the patient genotype . Since complete knockout of TXNL4A orthologs is lethal in multiple species, conditional or inducible knockout systems are essential for studying its function in specific tissues or developmental stages . CRISPR/Cas9-mediated homology-directed repair can introduce specific patient mutations into model organisms to study their functional consequences. For studying TXNL4A's role in cancer, genetically engineered mouse models with liver-specific TXNL4A overexpression, possibly in combination with other oncogenic drivers, could recapitulate aspects of hepatocellular carcinoma . Xenograft models using patient-derived HCC cells with manipulated TXNL4A expression levels would allow investigation of its effects on tumor growth, metastasis, and response to therapy. To study developmental aspects, researchers might employ zebrafish models, which offer advantages for visualizing craniofacial development in real-time. For any model, validation should include comprehensive assessment of splicing changes to confirm that the molecular consequences of TXNL4A manipulation resemble those observed in human patients. Additionally, phenotypic characterization should extend beyond gross morphology to include detailed analysis of relevant tissues and cell types, particularly neural crest cells for developmental models and immune infiltrates for cancer models .
TXNL4A contains a thioredoxin-like fold, which is a characteristic structural motif found in thioredoxin proteins . This fold allows TXNL4A to interact with multiple proteins, including the polyglutamine tract-binding protein 1 (PQBP1) . The primary function of TXNL4A is to participate in the assembly of the spliceosome, a complex responsible for removing introns from pre-mRNA .
TXNL4A plays a crucial role in the processing of capped intron-containing pre-mRNA and is involved in the mRNA splicing minor pathway . It is a component of the U5 snRNP and U4/U6-U5 tri-snRNP complexes, which are essential for the formation of the spliceosome . The protein’s interactions and functions are vital for the accurate and efficient splicing of pre-mRNA, ensuring proper gene expression and cellular function .
Mutations in the TXNL4A gene have been associated with Burn-McKeown syndrome, a rare genetic disorder characterized by craniofacial dysmorphisms, cardiac defects, hearing loss, and bilateral choanal atresia . Additionally, TXNL4A is linked to Developmental and Epileptic Encephalopathy 7, highlighting its importance in neurological development and function .
Recombinant forms of TXNL4A are used in research to study its structure, function, and interactions with other proteins. Understanding the role of TXNL4A in pre-mRNA splicing and its involvement in genetic disorders can provide insights into potential therapeutic targets for related diseases.