The protein binds GM-CSF with low affinity and forms a heterodimeric receptor with the beta subunit (CSF2RB) to activate downstream signaling pathways, including JAK-STAT and MAPK. Key roles include:
Functional Assays: Inhibits GM-CSF-dependent proliferation of TF1 erythroleukemic cells (ED₅₀ ≤ 10 µg/mL with 0.5 ng/mL GM-CSF) .
Disease Modeling: Used to study pulmonary alveolar proteinosis (PAP) and surfactant metabolism disorders linked to CSF2RA mutations .
DNAL4 (dynein axonemal light chain 4) is a protein encoded by the DNAL4 gene (also known as PIG27, MRMV3) located on chromosome 22q13.1. Despite its name suggesting axonemal function, research indicates DNAL4 plays a significant role in cytoplasmic dynein complexes involved in neurological development.
The protein appears crucial for netrin-1-directed retrograde transport, particularly in commissural neurons of the corpus callosum . This function is essential for proper brain wiring during development, as it facilitates axon guidance across the midline. Disruptions in this process can lead to aberrant neural connectivity, as observed in mirror movement disorders where voluntary movements on one side of the body are mirrored involuntarily on the opposite side .
Methodologically, researchers investigating DNAL4's developmental role often employ techniques such as immunohistochemistry on brain tissue sections, genetic knockout models, and protein interaction studies focused on netrin-1 signaling pathways to elucidate its precise functions in neural development.
DNAL4 mutations have been definitively linked to Mirror Movements 3 (MRMV3), a rare autosomal recessive disorder characterized by involuntary mirroring of voluntary movements between opposite sides of the body. The association was established through genetic analysis of a large consanguineous Pakistani family with 11 affected individuals across five generations .
Researchers employed the following methodological approaches to establish this link:
Initial linkage analysis and candidate gene approach (excluding previously implicated genes DCC and RAD51)
Microarray genotyping and autozygosity mapping, which identified a 3.3 Mb homozygous region on chromosome 22q13.1
Whole exome sequencing that identified a splice site mutation in DNAL4
Confirmation that this mutation leads to exon 3 skipping, resulting in a protein missing 28 amino acids
Linkage analysis using Simwalk2, yielding a maximum LOD score of 6.197, strongly supporting causality
The affected individuals exhibited classic mirror movement symptoms without other typical dynein-related phenotypes such as primary ciliary dyskinesis, situs inversus, or defective sperm, suggesting DNAL4's specialized role in corpus callosum development rather than in classic axonemal dynein functions .
DNAL4 is hypothesized to function within the cytoplasmic dynein complex specifically for netrin-1-directed retrograde transport in commissural neurons . This represents a specialized molecular mechanism distinct from DNAL4's potential role in axonemal dynein complexes.
The molecular pathway appears to involve:
Netrin-1 binding to its receptor DCC (deleted in colorectal cancer), which is also implicated in Mirror Movements 1 (MRMV1)
DNAL4-containing cytoplasmic dynein complexes mediating retrograde transport of netrin-1 signaling components
This transport facilitating proper axon guidance across the midline via the corpus callosum
Dysfunction leading to abnormal connectivity between left and right motor cortices
Researchers have observed that the splice site mutation in MRMV3 patients causes skipping of exon 3, resulting in an in-frame deletion of 28 amino acids. This suggests the mutant protein likely retains some function but has impaired interaction with other dynein complex components or cargo proteins involved in netrin-1 signaling .
Methodologically, protein interaction studies using techniques such as co-immunoprecipitation, yeast two-hybrid systems, and proximity ligation assays are valuable for elucidating DNAL4's binding partners in this pathway.
Several complementary experimental approaches have proven effective for investigating DNAL4:
CRISPR-Cas9 genome editing to create cell and animal models with DNAL4 mutations
siRNA and shRNA for gene silencing experiments to assess DNAL4 knockdown effects
Co-immunoprecipitation to identify interaction partners
Western blotting for protein expression level analysis
Immunohistochemistry to determine tissue and subcellular localization patterns
Live cell imaging with tagged DNAL4 to visualize retrograde transport
Axon guidance assays using microfluidic chambers with netrin-1 gradients
Neuronal migration assays in appropriate cell models
Mouse models with conditional Dnal4 knockout in specific neuronal populations
Examination of the rat Dnal4 ortholog, which has been annotated with "Mirror Movements 3" in the Rat Genome Database
The combination of these approaches allows researchers to investigate DNAL4 from molecular interactions to physiological functions in complex organisms.
DNAL4 belongs to the dynein light chain family but exhibits unique structural features that likely explain its specialized function in netrin-1-directed transport rather than classical axonemal roles.
Key structural aspects include:
The protein contains specific domains that facilitate integration into dynein complexes while maintaining cargo specificity
The 28 amino acids encoded by exon 3 (missing in MRMV3 patients) likely form a critical interaction surface
The protein appears to be evolutionarily conserved across species, suggesting functional importance
Interestingly, despite its name, DNAL4 (dynein axonemal light chain 4) likely functions primarily in cytoplasmic rather than axonemal dynein complexes in the context of neurological development. This highlights the importance of experimental validation over nomenclature-based assumptions .
Advanced structural biology techniques such as X-ray crystallography or cryo-electron microscopy of DNAL4 in complex with other dynein components would provide valuable insights into its precise structural role within these motor protein complexes.
DNAL4 shows specific expression patterns across human tissues that provide clues to its functional roles:
While comprehensive expression data from the search results is limited, available resources such as the Human Protein Atlas can be utilized to examine DNAL4 expression . Generally, dynein components show high expression in tissues with motile cilia or high transport requirements.
For neurological functions specifically, DNAL4 would be expected to show expression in developing commissural neurons of the corpus callosum, consistent with its role in mirror movement disorders .
Methodologically, researchers can investigate tissue-specific expression through:
RNA-seq and microarray data analysis
Quantitative PCR for mRNA expression
Immunohistochemistry with validated antibodies
Single-cell RNA sequencing to identify cell type-specific expression patterns in complex tissues like brain
Understanding expression patterns can guide hypothesis generation about potential functions in different tissues and cell types, beyond the established neurological role.
Despite DNAL4's nomenclature as an "axonemal" light chain, evidence suggests it functions importantly in cytoplasmic dynein complexes, particularly in neurological development . Researchers employ several approaches to distinguish these roles:
Subcellular localization studies: Immunofluorescence microscopy with co-localization markers for axonemes (e.g., acetylated tubulin) versus cytoplasmic dynein complexes
Functional assays: Examining effects on ciliary motility (axonemal function) versus intracellular transport (cytoplasmic function)
Protein interaction studies: Identifying binding partners specific to each complex type
Mutant phenotype analysis: Comparing DNAL4 mutations to classical axonemal dynein defects (e.g., primary ciliary dyskinesia) versus cytoplasmic dynein defects (e.g., neurological disorders)
The absence of typical axonemal dynein-related phenotypes (primary ciliary dyskinesia, situs inversus, defective sperm) in MRMV3 patients with DNAL4 mutations strongly suggests its primary role in these patients involves cytoplasmic rather than axonemal dynein function .
Comparative studies examining DNAL4's role across dynein-related processes can help clarify its functional specificity and potential dual roles in different cellular contexts.
Several gene editing approaches can effectively model DNAL4 mutations for research purposes:
Precise mutation introduction: Creating the exact splice site mutation found in MRMV3 patients to study its specific effects
Exon 3 deletion: Directly modeling the exon skipping observed in patients
Complete knockout: For loss-of-function studies to determine DNAL4's essential roles
Conditional knockout: Using Cre-lox systems to delete DNAL4 in specific cell types or developmental stages
Commercial siRNA, shRNA, and lentiviral particles targeting DNAL4 are available for gene silencing studies , which can:
Provide temporal control over DNAL4 downregulation
Allow titration of suppression levels
Be used in difficult-to-transfect cell types via lentiviral delivery
Newer base editing or prime editing technologies can introduce precise point mutations without double-strand breaks, potentially increasing editing efficiency for specific mutations.
The choice of approach depends on research goals, with CRISPR methods generally preferred for modeling specific disease mutations, while RNAi approaches may be more suitable for initial functional screening.
Several experimental systems can effectively model DNAL4-related disorders, particularly MRMV3:
Neuronal cell lines: SH-SY5Y or similar neuroblastoma lines can be modified to express mutant DNAL4
Primary neuronal cultures: From animal models with Dnal4 mutations
iPSC-derived neurons: Either from MRMV3 patients or engineered with DNAL4 mutations
Organoids: Brain organoids to study developmental effects in 3D culture
Mouse models: The mouse ortholog (Dnal4) shares high homology with human DNAL4, making it suitable for genetic manipulation studies
Rat models: Also appropriate given the annotation of Dnal4 with Mirror Movements 3 in the Rat Genome Database
Zebrafish: Useful for visualizing neuronal development and motor behaviors in transparent embryos
For mirror movement disorders specifically, these models should be assessed for:
Corpus callosum development abnormalities
Aberrant ipsilateral corticospinal projections
Bilateral motor cortex activation during unilateral movement attempts
Behavioral assays for unintended mirror movements
Effective modeling requires a combination of molecular, cellular, anatomical, and behavioral analyses to fully characterize the effects of DNAL4 dysfunction.
Investigating DNAL4's interactions with netrin-1 signaling requires specialized approaches focused on retrograde transport mechanisms:
Co-immunoprecipitation: To identify direct binding partners within the dynein complex and netrin-1 pathway
Proximity ligation assays: For detecting interactions in situ within cells
Yeast two-hybrid screening: To discover novel interaction partners
Protein complementation assays: For validating suspected interactions
Live-cell imaging: Using fluorescently tagged netrin-1 receptors and DNAL4 to visualize transport
Microfluidic chambers: For creating netrin-1 gradients and observing axon guidance responses
Quantum dot-labeled netrin-1: For single-molecule tracking during retrograde transport
Double mutant analysis: Combining DNAL4 mutations with mutations in netrin-1 pathway components (e.g., DCC receptor)
Rescue experiments: Testing whether wildtype DNAL4 can rescue defects in netrin-1 signaling components and vice versa
Since DNAL4 mutations and DCC (a netrin-1 receptor) mutations both cause mirror movement disorders , understanding their functional relationship is critical for elucidating the molecular pathways underlying proper commissural axon guidance.
DNAL4 is part of the dynein complex, which is responsible for the movement of cilia and flagella. The dynein complex generates force towards the minus ends of microtubules, a process that is essential for the motility of these cellular structures . The protein has ATPase activity, which means it can hydrolyze ATP to produce energy required for its motor functions .
The DNAL4 protein is expressed in tissues that contain motile cilia or flagella. This includes respiratory epithelial cells, where it plays a role in the movement of respiratory cilia, and sperm cells, where it is involved in the movement of sperm flagella . The expression of DNAL4 is crucial for the proper functioning of these motile structures.
The primary biological function of DNAL4 is to facilitate the movement of cilia and flagella. This movement is essential for various physiological processes, including the clearance of mucus and debris from the respiratory tract and the motility of sperm cells, which is necessary for fertilization .
Mutations in the DNAL4 gene have been associated with certain medical conditions. For example, defects in DNAL4 can lead to primary ciliary dyskinesia, a disorder characterized by chronic respiratory tract infections, reduced fertility, and other symptoms due to impaired ciliary function . Additionally, DNAL4 has been implicated in congenital mirror movement disorder, a condition where voluntary movements on one side of the body are mirrored by involuntary movements on the opposite side .
Recombinant DNAL4 protein is used in various research applications to study the function and regulation of dynein complexes. Understanding the role of DNAL4 in ciliary and flagellar motility can provide insights into the mechanisms underlying related disorders and contribute to the development of therapeutic strategies .