Putative transcription factor essential for axon growth and guidance in the central and peripheral nervous systems. It repels CNS axons from the midline by upregulating the expression of the midline repellent, Slit, and its receptor, Robo.
UniGene: Dm.11710
The Longitudinals lacking (lola) gene encodes a family of more than 20 transcription factor isoforms generated through alternative splicing in Drosophila melanogaster. All lola isoforms share an N-terminal BTB (Broad-Complex, Tramtrack and Bric-à-brac) dimerization domain that facilitates protein-protein interactions, while at least 17 isoforms contain unique C-terminal zinc finger (ZF) motifs involved in specific DNA binding .
Functionally, lola plays essential roles in multiple developmental processes including:
Axon growth and guidance in both central and peripheral nervous system development
Neural circuit formation and synapse development
Embryonic gonad formation and adult ovary morphogenesis
Regulation of programmed cell death during oogenesis
Maintenance and differentiation of germline and neural stem cells
All lola isoforms share common N-terminal exons encoding the BTB domain, which facilitates dimerization between different lola variants to form heterodimers in vivo. The C-terminal regions are encoded by alternative 3' exons, providing isoform specificity .
Most lola isoforms contain paired zinc finger motifs in their variant regions:
The first zinc finger typically follows a CCHC structure and functions primarily as a protein interaction domain
The second zinc finger generally has a C2H2 structure and serves as the DNA-binding domain
Isoform-specific structural variations significantly impact function. For example, isoforms A and L contain distinct zinc finger configurations that influence their DNA-binding specificities and target gene regulation .
Expression patterns of specific lola isoforms show temporal and spatial regulation:
| Isoform | Embryonic Expression | Larval Expression | Adult Expression | Notable Tissues |
|---|---|---|---|---|
| A | Moderate | Low | High | CNS, PNS, adult locomotor neurons |
| B | Low | Moderate | Moderate | Various tissues including CNS |
| D | Low | Variable | Moderate | Primarily neural tissues |
| L | High | High | High | Embryonic CNS, adult neural tissues |
Isoform L shows constitutive expression throughout development, while isoform A expression increases significantly in adult stages, particularly in locomotor neural circuits . Expression profiles correlate with developmental stage-specific functions, with lola-L being critical during early development, evident by its homozygous lethality when mutated .
For successful expression and purification of recombinant lola isoforms A, B, D, and L, researchers should consider the following methodological approach:
Expression System Selection:
For full-length isoforms: Insect cell-based systems (Sf9 or S2 cells) provide appropriate post-translational modifications
For isolated DNA-binding domains: Bacterial expression (E. coli BL21(DE3)) can yield sufficient protein for binding studies
Vector Design Considerations:
Include appropriate affinity tags (6xHis or GST) at the N-terminus to avoid interfering with C-terminal DNA-binding domains
For isoforms containing paired zinc fingers, supplement growth media with ZnCl₂ (50-100 μM) to ensure proper folding
Consider codon optimization for the expression system of choice
Protein Extraction and Purification:
Two-step purification approach combining affinity chromatography followed by size exclusion chromatography
Include reducing agents (DTT or β-mercaptoethanol, 1-5 mM) in all buffers to maintain zinc finger integrity
For isoforms A and L, which may form inclusion bodies in bacterial systems, implement denaturing purification followed by controlled refolding
Quality Control Assessments:
Verify protein integrity through Western blot analysis using isoform-specific antibodies
Confirm DNA-binding activity using electrophoretic mobility shift assays (EMSA) with known binding sequences
This approach has been successfully employed to characterize the DNA-binding specificities of various lola isoforms and their interactions with target gene enhancers .
Recent research has established effective CRISPR/Cas9 methodologies for generating isoform-specific lola mutations:
gRNA Design Strategy:
Design two distinct guide RNAs targeting the isoform-specific C-terminal exon
Select target sites that flank functional domains (e.g., zinc finger regions) to ensure complete loss of function
Validate gRNA specificity using appropriate in silico tools to minimize off-target effects
Delivery Method:
Establish transgenic fly lines expressing each pair of gRNAs under U6 promoters
Cross these lines with flies expressing Cas9 in the germline to generate heritable mutations
Mutation Screening:
Perform PCR screening using primers spanning the targeted region
Sequence verify deletions to confirm frameshift or loss of functional domains
Validate loss of isoform expression using qRT-PCR with isoform-specific primers
Rescue Experiments:
Generate genomic constructs expressing only the target isoform to verify phenotype specificity
For isoforms A and L, BAC-based rescue constructs containing the complete genomic region for the specific isoform are recommended
This methodology has successfully generated mutations for all 20 lola isoforms, revealing that five isoforms (including L) are essential for early development, while mutations in isoform A result in severe locomotion defects in adult flies .
To identify isoform-specific DNA binding sites and target genes, researchers should implement a multi-omics approach:
In Vitro Binding Site Determination:
Protein binding microarrays (PBMs) with recombinant isoform-specific DNA-binding domains
Systematic evolution of ligands by exponential enrichment (SELEX) followed by high-throughput sequencing
Results from these approaches revealed distinct binding preferences among isoforms, with isoform L recognizing GC-rich motifs
Genome-Wide Binding Profiling:
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using isoform-specific antibodies or tagged recombinant proteins
For isoforms A and L, CUT&RUN offers improved resolution with lower cell input requirements
Data analysis should incorporate motif discovery algorithms to identify isoform-specific binding sites
Target Gene Identification:
RNA-seq analysis comparing wild-type and isoform-specific mutant tissues
For isoform A, focus on adult locomotor neurons where it shows highest expression
For isoform L, embryonic nervous system tissues should be prioritized
Validation Methods:
Luciferase reporter assays using candidate enhancer regions
Directed mutagenesis of binding sites followed by in vivo enhancer activity assays
CRISPR interference (CRISPRi) targeting isoform-specific binding sites
This integrated approach has identified that isoform L regulates genes involved in early neural development, while isoform A predominantly regulates genes associated with locomotor functions .
Recombinant lola proteins, particularly those containing BTB domains, commonly experience aggregation during expression and purification. Effective solutions include:
Optimization of Expression Conditions:
Reduce expression temperature to 16-18°C
Employ auto-induction media rather than IPTG induction
For isoforms A and L, use solubility enhancing tags such as MBP or SUMO
Buffer Composition Modifications:
Include 5-10% glycerol to enhance stability
Add low concentrations (50-200 mM) of L-arginine to reduce aggregation
Optimize salt concentration (typically 300-500 mM NaCl) to maintain solubility
For zinc finger-containing isoforms, maintain 50-100 μM ZnCl₂ in all buffers
Purification Strategy Adjustments:
Implement gradient elution during affinity chromatography
Include size exclusion chromatography as a final polishing step
Consider on-column refolding for difficult-to-express isoforms
Storage Considerations:
Flash-freeze aliquots in liquid nitrogen
Store at concentrations below 1 mg/ml to prevent concentration-dependent aggregation
Include 1 mM DTT in storage buffers to maintain reduced state of cysteine residues
Implementation of these strategies has significantly improved the yield and homogeneity of recombinant lola isoforms, particularly for the structurally complex isoform L .
Researchers frequently encounter contradictory results when studying lola isoform functions across different experimental systems. To resolve these discrepancies:
Standardization of Genetic Backgrounds:
Maintain consistent genetic backgrounds when comparing isoform-specific mutants
Back-cross mutant lines at least 6 generations to eliminate background effects
Include multiple independent mutant alleles to confirm phenotypic observations
Temporal Control of Gene Function:
Implement temperature-sensitive GAL4/GAL80ts system for temporally controlled knockdown/overexpression
Use isoform-specific rescue constructs under heat-shock promoters for precise temporal control
This approach helped resolve contradictory results for isoform L, demonstrating distinct functions in embryonic versus adult tissues
Tissue-Specific Analysis:
Employ MARCM (Mosaic Analysis with a Repressible Cell Marker) to generate tissue-specific mutant clones
Use tissue-specific GAL4 drivers with UAS-RNAi constructs to achieve targeted knockdown
These approaches revealed that isoform A functions primarily in locomotor neurons, explaining discrepancies when analyzed in whole-animal studies
Resolution of Molecular Redundancy:
Generate combinatorial isoform mutants to address functional redundancy
Employ quantitative proteomic approaches to identify compensatory changes in other isoforms
Structure-function analysis with chimeric proteins to identify critical functional domains
These approaches have successfully resolved contradictory findings regarding isoform L's role in neural development versus adult neurogenesis .
Recombinant lola isoforms serve as powerful tools for dissecting transcriptional regulatory networks:
Protein Interaction Network Mapping:
BioID or proximity labeling approaches using recombinant lola isoforms as baits
Co-immunoprecipitation coupled with mass spectrometry to identify isoform-specific interactors
Yeast two-hybrid screening using the BTB domain versus isoform-specific C-terminal regions
These approaches identified distinct protein interaction networks for isoforms A and L
Transcriptional Complex Analysis:
Sequential ChIP (ChIP-reChIP) to identify co-regulatory factors at specific genomic loci
In vitro reconstitution of transcriptional complexes with recombinant components
Analysis of heterodimeric complex formation between different lola isoforms
| Isoform Combination | Complex Formation | Functional Output |
|---|---|---|
| A-L heterodimer | Efficient | Enhances target gene expression |
| A-B heterodimer | Limited | Minimal transcriptional effect |
| L-D heterodimer | Moderate | Repressive activity |
Enhancer Function Analysis:
CRISPR activation/interference studies targeting lola binding sites
High-throughput enhancer activity assays with systematic mutation of binding sites
Correlation of binding site architecture with transcriptional output
Integration with Epigenomic Data:
Analysis of chromatin accessibility changes (ATAC-seq) in isoform-specific mutants
Histone modification profiles at lola target genes
Correlation of DNA methylation patterns with isoform binding
Implementation of these approaches revealed that isoform L functions primarily as a transcriptional activator of neural development genes, while isoform A regulates genes involved in synapse formation and locomotor function .
While lola proteins are extensively studied in neural development, emerging research has revealed important roles in non-neural tissues:
Ovarian Development and Function:
Immune System Regulation:
Preliminary evidence suggests roles for lola in hemocyte development
Isoform A may regulate genes involved in cellular immunity
Research opportunities exist to characterize immune-specific transcriptional targets
Metabolic Regulation:
Emerging data indicate that certain lola isoforms may regulate metabolic genes
Potential cross-talk between neural and metabolic regulatory networks
Recombinant proteins could help identify metabolic enzyme genes directly regulated by specific isoforms
Aging and Lifespan Determination:
lola-O mutations affect lifespan, suggesting broader roles in aging
Investigation of isoform-specific effects on aging-related processes
Potential conservation of these functions in mammalian orthologs
Cancer Research Applications:
Human orthologs of lola (BTB-ZF family proteins) are implicated in various cancers
Recombinant Drosophila proteins serve as models for studying conserved mechanisms
Structure-function studies to inform therapeutic targeting of human orthologs
These emerging areas represent significant opportunities for researchers working with recombinant lola isoforms to expand our understanding beyond the well-established neural functions .
Evolutionary analysis provides valuable insights into lola isoform functions:
Comparative Genomics Approach:
Analysis of lola orthologs across Drosophila species reveals differential conservation patterns
The BTB domain shows high conservation (>90% identity), while C-terminal regions of isoforms show variable conservation
Isoforms L and A show higher sequence conservation than isoforms B and D, suggesting stronger evolutionary constraints
Structure-Function Correlation:
Mapping conserved residues onto protein structural models identifies functionally critical regions
For isoform L, zinc finger residues contacting DNA show nearly 100% conservation across Drosophila species
Variable regions between conserved motifs may contribute to species-specific functions
Mammalian Ortholog Analysis:
Human ZBTB family proteins represent functional orthologs of Drosophila lola
Cross-species rescue experiments using recombinant proteins can identify conserved functions
Sequence-structure-function comparative analysis informs translational research applications
Evolutionary Rate Analysis:
Calculation of dN/dS ratios for different protein domains reveals selective pressures
Higher evolutionary constraints on DNA-binding domains of isoforms A and L correlate with their essential functions
Sites under positive selection may indicate adaptation to species-specific regulatory requirements
This evolutionary approach has revealed that the DNA-binding specificity of isoform L is highly conserved across Diptera, while isoform A shows more rapid evolution, potentially contributing to species-specific locomotor behaviors .
Emerging technologies promise to revolutionize our understanding of lola isoform functionality:
Single-Cell Multi-Omics:
Single-cell RNA-seq combined with ATAC-seq to correlate isoform expression with chromatin accessibility
Single-cell proteomics to detect isoform-specific protein expression patterns
Spatial transcriptomics to map isoform expression within complex tissues
These approaches will reveal cell-type specific functions of different isoforms that may be masked in bulk analyses
CRISPR-Based Functional Genomics:
Prime editing for precise introduction of isoform-specific mutations
CRISPR activation/interference screens targeting putative enhancers
Base editing to introduce specific amino acid changes in zinc finger domains
These techniques will allow fine-grained analysis of structure-function relationships
Cryo-EM and Structural Biology:
Determination of isoform-specific protein complex structures
Analysis of DNA-bound conformations of different isoforms
Structural basis for heterodimer formation between isoforms
Structural insights will inform rational design of isoform-specific inhibitors or activators
Systems Biology Integration:
Multi-scale modeling of transcriptional networks
Integration of transcriptomic, proteomic, and metabolomic data
Machine learning approaches to predict isoform-specific functions from sequence
These integrative approaches will place isoform-specific functions within broader biological contexts
These technological advances will help resolve current knowledge gaps, particularly regarding the functions of less-characterized isoforms B and D, and potential synergistic or antagonistic relationships between different isoforms .
Recombinant lola proteins represent valuable tools for translational neuroscience research:
Disease-Associated Variant Modeling:
Human BTB-ZF family proteins related to lola are implicated in neurodevelopmental disorders
Introduction of disease-associated mutations into recombinant lola proteins allows functional characterization
Transcriptional output changes can be measured using reporter assays
This approach has identified potential disease mechanisms for mutations in human ZBTB proteins
Drug Discovery Applications:
High-throughput screening platforms using recombinant lola proteins
Identification of small molecules that modulate isoform-specific DNA binding
Structure-based drug design targeting the BTB domain to disrupt protein-protein interactions
These approaches may yield lead compounds for neurological disorder therapies
Gene Therapy Development:
Design of engineered transcription factors based on lola DNA-binding domains
Targeted gene regulation for neurodevelopmental disorders
Optimization of delivery methods using Drosophila as a model system
Proof-of-concept studies demonstrate feasibility of this approach for regulating disease-associated genes
Biomarker Discovery:
Identification of downstream targets of lola with conserved regulation in humans
Development of diagnostic panels based on transcriptional signatures
Correlation of gene expression patterns with disease progression
These translational applications highlight the value of basic research on lola isoforms for understanding and potentially treating human neurological disorders with dysregulated transcriptional control .