Recombinant Mouse LEM domain-containing protein 2 (Lemd2) is a full-length protein derived from the mouse gene Lemd2. This protein is part of the inner nuclear membrane (INM) and plays a crucial role in maintaining nuclear structure and regulating signaling pathways. The recombinant form of Lemd2 is typically expressed in Escherichia coli (E. coli) and is often used in research to study its functions and interactions within the cell.
Lemd2 is a transmembrane protein with a molecular weight of approximately 58 kDa in its native form. It contains a LEM domain, which is a characteristic motif found in proteins associated with the nuclear lamina, such as emerin and MAN1. The LEM domain facilitates interactions with barrier-to-autointegration factor (BAF), contributing to chromatin organization and nuclear envelope integrity .
Species: Mus musculus (Mouse)
Source: E. coli
Tag: His-tagged for purification
Protein Length: Full-length (511 amino acids)
Form: Lyophilized powder
Lemd2 is involved in several critical biological processes:
Nuclear Structure and Integrity: It contributes to maintaining the organization of the nuclear envelope and interacts with lamin A/C and other nuclear lamina proteins .
Signaling Pathways: Lemd2 regulates key signaling cascades, including the mitogen-activated protein kinase (MAPK) and protein kinase B (AKT) pathways. Its absence can lead to enhanced activation of these pathways, which is associated with various cellular dysfunctions .
Embryonic Development: Studies in mice have shown that Lemd2 is essential for embryonic development, with its deficiency leading to embryonic lethality around E11.5 .
Research on Lemd2 has provided insights into its role in health and disease:
Muscle Differentiation: Lemd2 promotes myoblast differentiation by attenuating ERK signaling, highlighting its importance in muscle development .
Disease Models: Mutations in LEMD2 have been linked to progeroid syndromes and other nuclear envelopathies, emphasizing its potential as a candidate gene for human diseases .
Cardiac Implications: Studies have shown that alterations in Lemd2 function can lead to severe cardiac abnormalities, such as dilated cardiomyopathy .
Feature | Description |
---|---|
Species | Mus musculus (Mouse) |
Source | E. coli |
Tag | His-tagged |
Length | Full-length (511 amino acids) |
Form | Lyophilized powder |
Purity | Greater than 90% |
Lemd2 is a novel LEM domain protein structurally related to MAN1. It contains an N-terminal LEM motif, two predicted transmembrane domains, and a MAN1-Src1p C-terminal (MSC) domain highly homologous to MAN1. Unlike MAN1, Lemd2 lacks the MAN1-specific C-terminal RNA-recognition motif. The LEM domain mediates binding to barrier-to-autointegration factor (BAF), a DNA-crosslinking protein . Lemd2's structural composition facilitates its function as a lamina-associated protein residing in the inner nuclear membrane (INM), where it interacts with the nuclear lamina.
Lemd2 is primarily localized to the inner nuclear membrane (INM). Immunofluorescence microscopy of digitonin-treated cells and subcellular fractionation has identified Lemd2 as a lamina-associated protein residing in the INM . Targeting of Lemd2 to the nuclear envelope requires A-type lamins and is mediated by the N-terminal and transmembrane domains. When investigating Lemd2 localization, researchers should consider using:
Digitonin permeabilization to distinguish INM from outer nuclear membrane proteins
Co-localization with known nuclear lamina markers
Subcellular fractionation to biochemically verify membrane association
Lemd2 has been shown to:
The interaction between Lemd2 and SATB2 is particularly significant as it links nuclear shape plasticity to neuronal activity-dependent gene regulation .
For optimal visualization of Lemd2's subcellular localization, researchers should employ a multi-method approach:
Immunofluorescence with selective permeabilization:
Use digitonin (0.001-0.005%) for selective permeabilization of the plasma membrane but not nuclear membranes
Follow with antibodies against Lemd2 and other nuclear envelope markers (lamin A/C, emerin)
Include controls with Triton X-100 permeabilization to access all cellular compartments
Subcellular fractionation:
Separate nuclear envelope fractions from other cellular components
Perform Western blot analysis using antibodies against Lemd2 and control proteins
Include markers for different nuclear compartments (INM, ONM, nuclear lamina)
Electron microscopy with immunogold labeling:
For the highest resolution localization within the nuclear envelope
These approaches should be used complementarily to verify consistent localization patterns.
Based on published research methodologies, effective Lemd2 depletion can be achieved through:
siRNA transfection:
shRNA for stable knockdown:
More suitable for long-term experiments
Can be delivered via lentiviral vectors for hard-to-transfect cells
CRISPR-Cas9 genome editing:
For complete knockout studies or introduction of specific mutations
Can be designed to target early exons of Lemd2
Each approach should include appropriate controls and validation of knockdown/knockout efficiency at both mRNA and protein levels.
Production of high-quality recombinant mouse Lemd2 requires careful consideration of its membrane protein nature:
Expression systems:
Bacterial expression (E. coli): Best for soluble domains (LEM domain, C-terminal region)
Insect cell expression (Sf9, High Five): More suitable for full-length protein with proper folding
Mammalian expression (HEK293, CHO): Optimal for fully functional protein with mammalian post-translational modifications
Purification strategy:
For full-length protein: Detergent solubilization (DDM, CHAPS, or digitonin)
Affinity tags: His6, GST, or FLAG tags positioned to avoid interference with protein function
Size exclusion chromatography for final purity
Quality control assessments:
SDS-PAGE and Western blot analysis
Mass spectrometry for identity confirmation
Circular dichroism to verify proper folding
Functional binding assays with known partners (lamin C, BAF)
Lemd2 plays a crucial role in activity-dependent nuclear envelope plasticity through its interaction with SATB2. Research has shown that:
Neuronal activation through action potential bursting leads to changes in nuclear geometry
Both SATB2 and Lemd2 are required for this nuclear envelope remodeling
The process also involves the ESCRT-III/VPS4 membrane-remodeling complex
When investigating this phenomenon, researchers should:
Use physiologically relevant neuronal activation paradigms (e.g., bicuculline treatment in vitro)
Apply advanced imaging techniques to capture nuclear shape changes
Consider temporal dynamics of the remodeling process
Include appropriate controls for neuronal activity states
Lemd2 depletion has profound effects on gene expression in cortical neurons, particularly under conditions of neuronal activation. Key findings include:
In bicuculline-treated (synaptically active) neuronal cultures, Lemd2 knockdown results in:
The effect is much weaker in NBQX-treated (inactive) neurons, indicating Lemd2 is particularly relevant for gene regulation in active neurons
Immediate early response (IEG) genes are particularly affected:
Gene Expression Effects | Lemd2 Knockdown | SATB2 Knockout | Overlap |
---|---|---|---|
All Primary Response Genes | 50/135 down-regulated | 58/135 down-regulated | 26 genes |
Early Response Genes | 53/103 down-regulated | 41/103 down-regulated | 31 genes |
This data demonstrates significant overlap between Lemd2- and SATB2-regulated genes, suggesting they function in the same pathway to regulate activity-dependent gene expression .
Compelling genetic evidence connects Lemd2-regulated genes to neuropsychiatric disorders and cognitive function:
Common genetic variants: Lemd2-regulated genes are significantly enriched for common variants associated with:
Rare de novo mutations: Lemd2-regulated genes are enriched for:
Control analyses verify specificity:
No enrichment for synonymous mutations in these disorders
No enrichment in control trios or unaffected siblings
No enrichment for eight other tested phenotypes (childhood-onset psychiatric disorders, other brain-related diseases, and non-brain-related diseases)
Enrichment remains significant even after controlling for "brain-expressed" and "brain-elevated" gene sets
These findings suggest that Lemd2-regulated gene networks contribute to human cognitive function and neuropsychiatric disorder risk, similar to the genes regulated by its interaction partner SATB2 .
To effectively investigate the interaction between Lemd2 and SATB2, researchers should employ multiple complementary techniques:
Co-immunoprecipitation (Co-IP):
Use antibodies against endogenous proteins where possible
Include appropriate negative controls and non-interacting protein controls
Consider crosslinking for transient interactions
Validate results in both directions (IP Lemd2, blot for SATB2 and vice versa)
Proximity ligation assay (PLA):
Provides spatial information about protein interactions in situ
Can detect endogenous protein interactions at single-molecule resolution
Particularly useful for visualizing interactions at the nuclear envelope
FRET/BRET approaches:
For studying interaction dynamics in living cells
Requires fusion proteins with appropriate fluorophore/luminescent tags
Can provide quantitative measures of interaction strength
Protein fragment complementation assays:
Split GFP or split luciferase systems can confirm direct interactions
Useful for screening interaction domains
Protein domain mapping:
Generate truncation or deletion mutants to identify specific interaction domains
Point mutations can identify critical residues for interaction
Visualizing and quantifying nuclear shape changes requires sophisticated imaging and analysis approaches:
Live cell imaging techniques:
Fluorescent protein tagging of nuclear envelope markers (e.g., lamin B1-GFP)
Spinning disk or light sheet microscopy for reduced phototoxicity during long-term imaging
Time-lapse imaging at appropriate intervals (1-5 minutes) to capture dynamics
Super-resolution microscopy:
STED, STORM, or PALM for nanoscale resolution of nuclear envelope structure
Can be combined with expansion microscopy for enhanced resolution
Quantitative image analysis:
Nuclear morphometric analysis (NMA) to quantify shape parameters
Measure nuclear invagination frequency, depth, and distribution
Fourier shape descriptors to characterize complex morphological changes
3D reconstruction for comprehensive morphological assessment
Activity manipulation protocols:
Experimental design considerations:
Include appropriate controls (Lemd2 knockdown/knockout, SATB2 deficient)
Consider temporal dynamics (immediate vs. sustained changes)
Correlate morphological changes with functional readouts (gene expression)
Based on published research methodologies, the following transcriptomic approaches are most informative:
RNA-seq experimental design:
Compare Lemd2-depleted vs. control neurons under different activity states:
Include appropriate controls for neuronal activity effects
Use sufficient biological replicates (minimum n=3) for statistical power
Differential expression analysis:
Functional annotation approaches:
Gene Ontology (GO) analysis to identify enriched functional categories
Pathway analysis (KEGG, Reactome) for biological context
Focus on neuron-specific pathways and processes
Integration with genomic data:
Validation approaches:
qRT-PCR validation of key differentially expressed genes
Protein-level validation when possible
Rescue experiments to confirm specificity
This multi-layered approach has successfully identified Lemd2's role in regulating immediate early genes and developmental transcription factors in neurons .
Detecting endogenous Lemd2 can be challenging due to several factors:
Antibody specificity issues:
Solution: Validate antibodies using Lemd2 knockdown/knockout controls
Use multiple antibodies targeting different epitopes
Consider epitope-tagged knock-in models for difficult detection scenarios
Low expression levels in certain cell types:
Solution: Optimize protein extraction protocols with nuclear envelope enrichment
Increase loading amounts for Western blots
Use signal enhancement systems for immunofluorescence
Masked epitopes due to protein-protein interactions:
Solution: Test multiple fixation and permeabilization conditions
Try antigen retrieval methods
Consider native vs. denaturing conditions for Western blotting
Cross-reactivity with other LEM domain proteins:
Solution: Include appropriate controls (other LEM protein knockdowns)
Perform peptide competition assays to confirm specificity
Use mass spectrometry for definitive identification
Sample preparation artifacts affecting nuclear envelope structure:
Solution: Compare multiple fixation methods
Validate findings with live cell imaging when possible
Include appropriate controls for each preparation method
Distinguishing direct from indirect effects of Lemd2 depletion requires a multi-faceted experimental approach:
Temporal analysis:
Perform time-course experiments after Lemd2 depletion
Early changes (hours) are more likely to be direct effects
Late changes (days) may represent secondary effects
Correlation with chromatin interactions:
Rescue experiments:
Re-express Lemd2 after knockdown to identify reversible changes
Use domain mutants to identify regions necessary for specific effects
Design rapid inducible/degradable systems for temporal control
Combined genomic approaches:
Integrate RNA-seq with ATAC-seq to correlate expression changes with chromatin accessibility
Hi-C or Chromosome Conformation Capture to detect changes in chromatin organization
Nuclear run-on assays (e.g., PRO-seq) to measure nascent transcription directly
Computational approaches:
Network analysis to identify primary nodes vs. downstream effects
Causal inference algorithms to predict direct regulatory relationships
Comparison with published datasets of transcription factor knockdowns
The current evidence suggests that Lemd2's effects on immediate early genes are likely direct, as they occur rapidly upon neuronal activation and many affected genes contain SATB2 binding sites in their promoters .
When investigating Lemd2's role in nuclear shape dynamics, these critical controls should be included:
Protein level controls:
Validate Lemd2 knockdown/knockout efficiency by Western blot and qPCR
Include scrambled siRNA or appropriate negative controls
Rescue experiments with wild-type Lemd2 to confirm specificity
Activity manipulation controls:
Imaging controls:
Blind analysis to prevent observer bias
Include wild-type controls processed in parallel
Use multiple nuclear envelope markers to verify consistent effects
Include non-neuronal cells as negative controls for neuron-specific effects
Pathway component controls:
Temporal controls:
Establish baseline nuclear morphology before stimulation
Include multiple time points to capture dynamics
Recovery period after stimulation to assess reversibility
By implementing these controls, researchers can more confidently attribute observed nuclear shape changes to Lemd2's function rather than to experimental artifacts or indirect effects.
Given the links between Lemd2-regulated genes and neuropsychiatric disorders, several promising therapeutic approaches emerge:
Small molecule screening:
Develop high-throughput assays for SATB2-Lemd2 interaction
Screen for compounds that modulate this interaction
Test effects on neuronal activity-dependent gene expression
Gene therapy approaches:
CRISPR activation/interference systems targeting Lemd2 or key regulated genes
Viral delivery of modified Lemd2 variants to restore function in models of dysfunction
AAV-based approaches for CNS delivery
Peptide-based therapeutics:
Design peptides that mimic interaction domains
Cell-penetrating peptides to target nuclear envelope interactions
Stapled peptides for enhanced stability and cellular uptake
Preclinical model development:
Generate conditional Lemd2 knockout mouse models
Develop human iPSC-derived neuronal models with Lemd2 mutations
Establish behavioral assays relevant to cognitive function and psychiatric symptoms
Biomarker development:
Identify accessible biomarkers of Lemd2 pathway dysfunction
Develop imaging approaches to assess nuclear envelope dynamics in vivo
Correlate with clinical measures of cognitive function
Therapeutic development should focus on restoring normal activity-dependent gene regulation rather than constitutive activation of this pathway.
Single-cell technologies offer powerful new avenues for Lemd2 research:
Single-cell RNA-seq applications:
Identify cell type-specific effects of Lemd2 depletion
Explore heterogeneity in responses to neuronal activation
Map developmental trajectories affected by Lemd2 dysfunction
Correlate with spatial information (Spatial transcriptomics)
Single-cell ATAC-seq or CUT&TAG:
Map chromatin accessibility changes at single-cell resolution
Identify regulatory elements affected by Lemd2-SATB2 interaction
Correlate with gene expression changes
Single-cell imaging approaches:
Live imaging of nuclear dynamics in individual neurons
Correlate morphological changes with functional outcomes
Track long-term changes in individual cells after stimulation
Single-cell multi-omics:
Combined RNA-seq and ATAC-seq from the same cells
Protein and RNA co-detection to correlate Lemd2 levels with gene expression
Chromosome conformation with gene expression
Computational integration:
Trajectory inference to map cellular states during activation
Network analysis at single-cell level
Integration with human genetic data at cell-type specific resolution
These approaches would help resolve cell-type specific functions of Lemd2 that may be obscured in bulk tissue analyses.
The Lemd2-SATB2 interaction may represent an evolutionarily significant mechanism for cognitive function:
Evolutionary conservation analysis:
The interaction domains of Lemd2 and SATB2 show high conservation across vertebrates
The regulatory network they control includes many genes with human-specific features
Many Lemd2-regulated genes show accelerated evolution in human lineage
Comparative genomics implications:
Developmental timing significance:
The activity-dependent nuclear remodeling may contribute to critical period plasticity
This mechanism could underlie experience-dependent wiring during development
Evolutionary changes in this pathway might relate to extended human cognitive development
Theoretical framework:
Nuclear envelope-chromatin interactions represent a physical basis for experience-dependent gene regulation
This physical regulation may allow for more complex integration of signals
Evolution of nuclear architecture regulation may parallel cognitive complexity across species
Further comparative studies across species with varying cognitive capabilities would help elucidate the evolutionary significance of this pathway.