DHRS4 catalyzes the reduction of carbonyl-containing substrates with stereochemical specificity:
Substrate | Product | Stereochemistry | Activity Level |
---|---|---|---|
3-Keto-C19/C21 Steroids | 3β-Hydroxysteroids | Human-specific | High |
Benzil | R-Benzoin | Human-specific | High |
All-trans-Retinal | All-trans-Retinol | Low | Low |
9,10-Phenanthrenequinone | Reduced metabolites | Not specified | Moderate |
Retinoid Metabolism: Converts retinal to retinol, competing with retinoic acid (ATRA) synthesis. Upregulation of DHRS4 in ALS models reduces ATRA levels, impairing neuroprotective signaling .
Xenobiotic Detoxification: Reduces cytotoxic quinones (e.g., 9,10-phenanthrenequinone) but does not enhance cytotoxicity in transfected cells .
DHRS4 overexpression is implicated in neurodegenerative diseases:
Retinoid Imbalance: DHRS4-mediated reduction of retinal to retinol diverts precursors from ATRA synthesis, reducing neuroprotective signaling .
Immune Dysregulation: Upregulation correlates with neuroinflammation, including macrophage activation and synapse pruning pathways .
DHRS4 exhibits broad tissue distribution with specialized roles:
Tissue | Expression Level | Key Functions | Source |
---|---|---|---|
Brain | Moderate | Retinoid metabolism, neuroprotection | |
Liver | High | Steroid metabolism, PPARα regulation | |
Endothelial Cells | Inducible | Steroid hormone synthesis |
Recombinant DHRS4 is utilized in:
Enzymatic Assays: Studying NADPH-dependent reductase activity .
Disease Modeling: Investigating ALS pathogenesis and retinoid signaling .
Xenobiotic Studies: Evaluating detoxification mechanisms for aromatic ketones and quinones .
Induction: Upregulated by peroxisome-proliferator-activated receptor α (PPARα) ligands in hepatic cells .
Storage: Requires carrier proteins (e.g., BSA) for long-term stability .
Human DHRS4 differs from non-primate orthologs in:
DHRS4 functions primarily as a reductase that catalyzes the conversion of all-trans-retinal and 9-cis retinal to their corresponding retinol forms. It can also catalyze the reverse reaction (oxidation of all-trans-retinol) when utilizing NADP as a cofactor, though with lower efficiency. Additionally, it reduces alkyl phenyl ketones and alpha-dicarbonyl compounds with aromatic rings, including pyrimidine-4-aldehyde, 3-benzoylpyridine, 4-benzoylpyridine, menadione, and 4-hexanoylpyridine. Notably, DHRS4 shows no activity toward aliphatic aldehydes and ketones .
DHRS4 belongs to the short-chain dehydrogenases/reductases (SDR) family, sharing structural and functional similarities with DHRS3 and DHRS9. These proteins all participate in retinoid metabolism, with overlapping yet distinct substrate specificities. DHRS3 and DHRS4 show particularly high functional overlap, as both catalyze the reduction of all-trans retinal to all-trans retinol in the presence of NADPH . This functional redundancy suggests potential compensatory mechanisms within biological systems, which should be considered when designing gene knockout experiments.
For robust detection of DHRS4 expression, a multi-platform approach is recommended:
Western blotting: Effective for quantifying protein levels in tissue lysates
Immunofluorescence: Useful for visualizing cellular localization (primarily cytoplasmic in neurons)
qRT-PCR: For mRNA expression analysis
When performing immunofluorescence, co-staining with neuronal markers like NeuN can help identify cell-specific expression patterns. For example, in SOD1^G93A ALS mouse models, DHRS4 showed increased cytoplasmic expression in neurons compared to wild-type mice . For protein extraction from spinal cord tissue, homogenization in RIPA buffer with protease inhibitors, followed by centrifugation at 12,000g for 15 minutes at 4°C, yields optimal results for subsequent Western blot analysis.
WGCNA is a powerful bioinformatics approach for identifying gene modules correlated with DHRS4 expression or disease progression. When applying WGCNA to DHRS4 research, consider the following methodological steps:
Data preprocessing: Normalize expression data and filter low-quality samples
Network construction: Select an appropriate soft-thresholding power to achieve scale-free topology
Module identification: Use hierarchical clustering and dynamic tree cutting
Correlation analysis: Correlate module eigengenes with clinical traits (e.g., DHRS4 expression, disease status)
In ALS research, WGCNA identified modules where DHRS4 was positively correlated with disease progression. The red module in GSE52946 data showed significant correlation with both DHRS4 expression and ALS patient status . This approach revealed DHRS4's potential association with immune-related pathways, such as neutrophil degranulation and macrophage activation.
Validating DHRS4 as a biomarker requires a comprehensive multi-stage approach:
Discovery phase:
Differential expression analysis across disease stages
ROC curve analysis to determine diagnostic potential
Correlation with clinical parameters
Validation phase:
Cross-validation in multiple independent cohorts
Longitudinal studies tracking expression changes
Comparison with established biomarkers
Functional validation:
Knockdown/overexpression studies
Pathway analysis of downstream effects
In ALS research, DHRS4 validation included analysis across multiple datasets (GSE52946, GSE10953, GSE43879, GSE46298) showing consistent upregulation with disease progression . Experimental validation using Western blotting and immunofluorescence confirmed these findings. ROC curve analysis demonstrated high diagnostic value (AUC>0.7) when combined with related genes (C1Q complex, C3, ITGB2) .
PPI network analysis for DHRS4 should follow these methodological steps:
Data collection:
Extract DHRS4 co-expressed genes from relevant modules
Include known interactors from databases like STRING
Network construction:
Upload gene lists to STRING database (http://www.string-db.org/)
Set appropriate confidence score thresholds (recommended: 0.4-0.7)
Include experimental and database interaction sources
Network analysis:
Identify central nodes through centrality measures
Perform cluster analysis to identify functional modules
Conduct enrichment analysis of network components
In DHRS4 research, PPI analysis revealed interactions with complement cascade components (C1QA, C1QB, C1QC, C3) and integrin subunit beta 2 (ITGB2), suggesting immune-mediated functions . The network visualization showed these components occupying central positions, highlighting their potential importance in DHRS4-related pathways.
Multiple lines of evidence support DHRS4 as a potential biomarker for ALS progression:
Transcriptomic evidence:
Significant upregulation in ALS patient spinal cord compared to healthy controls
Progressive increase in expression with disease advancement in SOD1^G93A mice
Consistent findings across multiple independent datasets (GSE52946, GSE10953, GSE43879, GSE46298)
Proteomic evidence:
Increased protein expression verified by Western blotting
Enhanced immunoreactivity in motor neurons of ALS models
Functional relevance:
Association with complement cascade activation
Correlation with immune cell infiltration
Experimental validation showed that DHRS4 expression increases from pre-onset (60-70 days) through onset (90-100 days) to progression (120-130 days) stages in SOD1^G93A mice, paralleling the clinical course of disease . This temporal pattern of expression makes DHRS4 particularly valuable as a progression rather than simply a diagnostic biomarker.
DHRS4's interaction with the immune system in neurodegenerative diseases involves multiple mechanisms:
Methodologically, these associations were established through immune infiltration analysis using the ImmuCellAI tool (http://bioinfo.life.hust.edu.cn/web/ImmuCellAI/), followed by Spearman's correlation analysis between DHRS4 expression and immune cell abundance estimates . This suggests that monitoring DHRS4 expression may provide insights into neuroimmune interactions in disease states.
For studying DHRS4 in neurodegeneration, multiple complementary models are recommended:
In vivo models:
SOD1^G93A transgenic mice (most validated for ALS studies)
Age-matched cohorts at pre-onset, onset, and progression stages
Conditional DHRS4 knockout/overexpression models
In vitro models:
Primary motor neuron cultures
iPSC-derived motor neurons from patients and controls
Microfluidic chambers for axonal transport studies
Ex vivo models:
Organotypic spinal cord slices
Patient-derived tissue samples
The SOD1^G93A mouse model provides several advantages for DHRS4 research, including well-characterized disease progression, similar pathology to human ALS, and the ability to study pre-symptomatic stages . When using this model, proper genotyping via PCR of tail DNA is essential, and standard housing conditions (12h light/dark cycle, 20°C–27°C, 40%–50% humidity) should be maintained to ensure reproducibility.
DHRS4 interacts with several key protein partners that contribute to its biological functions:
Interaction Partner | Interaction Score | Functional Relevance |
---|---|---|
ALDH1A1 | 0.963 | Retinal oxidation to retinoic acid |
DHRS9 | 0.949 | 3-alpha-hydroxysteroid dehydrogenase activity |
RETSAT | 0.947 | All-trans-retinol saturation |
DHRS3 | N/A (Synergistic) | Catalyzes reduction of all-trans retinal |
C1QA/B/C | N/A (Disease context) | Complement cascade components |
C3 | N/A (Disease context) | Central complement component |
To study these interactions effectively:
Co-immunoprecipitation (Co-IP): Use anti-DHRS4 antibodies to pull down protein complexes
Proximity ligation assay (PLA): For detecting protein-protein interactions in situ
Bimolecular fluorescence complementation (BiFC): For visualizing direct interactions
FRET/FLIM: For analyzing dynamic interactions in living cells
Validation should include both overexpression systems and endogenous protein detection to avoid artifacts. When studying the DHRS4-DHRS3 synergistic relationship, consider analyzing both proteins simultaneously, as they share functional overlap in retinoid metabolism .
DHRS4 plays a multifaceted role in retinoid metabolism:
Primary function: Reduces all-trans-retinal and 9-cis retinal to their corresponding retinol forms
Secondary function: Can oxidize all-trans-retinol with NADP as cofactor (lower efficiency)
Pathway context: Works alongside DHRS3, DHRS9, and ALDH1A1 in retinoid interconversion
For studying DHRS4's role in retinoid metabolism, these methodologies are recommended:
Enzymatic activity assays:
Spectrophotometric measurement of NAD(P)H consumption
HPLC analysis of retinoid conversion
LC-MS/MS for comprehensive retinoid profiling
Cell-based assays:
Retinoid-responsive reporter gene assays
Metabolic labeling with deuterated retinoids
Live-cell imaging with fluorescent retinoid analogs
In silico approaches:
Molecular docking to predict substrate binding
Pathway flux analysis for system-level understanding
When designing experiments, consider that DHRS4 shows substrate specificity for aromatic aldehydes and ketones but not aliphatic compounds . Control experiments should include both positive controls (known substrates like all-trans-retinal) and negative controls (aliphatic aldehydes).
Single-cell transcriptomics offers powerful new approaches for DHRS4 research:
Cell type-specific expression:
Identification of DHRS4-expressing cell populations
Characterization of expression heterogeneity within cell types
Correlation with cell state markers
Methodological considerations:
Tissue dissociation protocols that preserve cellular integrity
FACS-based enrichment of neuronal populations
Computational analysis pipelines sensitive to low-abundance transcripts
Integration with spatial transcriptomics:
Combined single-cell and spatial analysis to map DHRS4-expressing cells
Correlation with anatomical features and pathological hallmarks
Current evidence suggests DHRS4 is predominantly expressed in neuronal cytoplasm in the spinal cord . Single-cell approaches could reveal whether specific neuronal subtypes (e.g., motor neurons vs. interneurons) show differential expression or regulation, potentially explaining selective vulnerability in diseases like ALS.
Developing therapeutic approaches targeting DHRS4 faces several challenges:
Target validation challenges:
Determining whether DHRS4 upregulation is causative or reactive in disease
Understanding compensatory mechanisms involving other SDR family members
Clarifying tissue-specific roles and potential off-target effects
Methodological approaches:
Conditional knockout models to assess temporal requirements
Pharmacological inhibition with selective compounds
AAV-mediated gene therapy approaches
Translational considerations:
Development of blood-based assays for monitoring DHRS4 activity
Identification of DHRS4 pathway modulators with BBB penetrance
Establishment of human iPSC-based screening platforms
The overlapping functions of DHRS4 with DHRS3 and other SDR family members present both a challenge and opportunity - while redundancy may limit efficacy of single-target approaches, it also suggests combination strategies targeting multiple pathway components might be more effective .
Computational approaches offer powerful tools for predicting novel DHRS4 functions:
Structure-based approaches:
Homology modeling based on SDR family crystal structures
Molecular dynamics simulations to identify conformational changes
Virtual screening for novel substrates and inhibitors
Network-based predictions:
Network expansion beyond first-degree interactions
Inference of indirect functional associations through guilt-by-association
Identification of disease modules containing DHRS4
Implementation methodology:
Begin with high-confidence protein structure prediction using AlphaFold2
Perform substrate docking using AutoDock Vina or similar tools
Validate predictions with focused biochemical assays
Current network analyses have already identified unexpected connections between DHRS4 and complement cascade components . Expanding these approaches could reveal additional non-canonical functions beyond retinoid metabolism, potentially explaining DHRS4's role in complex diseases like ALS where multiple pathways may be dysregulated simultaneously.
The DHRS4 gene is located on chromosome 14q11.2 and encodes a protein that is 278 amino acids long . The protein is expressed in various tissues and is involved in several metabolic pathways, including retinoic acid signaling and peroxisomal lipid metabolism . The gene cluster also includes a paralog, DHRS4L2, which shares similar functions .
DHRS4 is an NADPH-dependent oxidoreductase, meaning it uses NADPH as a cofactor to catalyze the reduction of its substrates . The enzyme is particularly efficient in reducing all-trans-retinal and 9-cis retinal, which are forms of vitamin A . It can also catalyze the oxidation of all-trans-retinol, although with much lower efficiency . This activity is crucial for maintaining the balance of retinoids in the body, which are essential for vision, growth, and cellular differentiation.
Recombinant DHRS4 protein is widely used in biochemical research to study its enzymatic properties and potential therapeutic applications. The recombinant form is typically expressed in Escherichia coli and purified to high levels of purity for use in various assays, including SDS-PAGE and mass spectrometry .
Alterations in DHRS4 expression or function can have significant implications for human health. For instance, dysregulation of retinoid metabolism can lead to vision problems, skin disorders, and impaired immune function. Therefore, understanding the role of DHRS4 in these processes is essential for developing targeted therapies.