Primary Neurons RNA Silencing Guide
Transfection-free gene knockdown that preserves neuronal electrophysiology

Why Primary Neurons?
AUMsilence self-delivering antisense oligonucleotides (sdASOs) enable transfection-free gene silencing through receptor-mediated endocytic pathways . Cellular uptake occurs through multiple endocytic pathways facilitated by phosphorothioate backbone modifications that promote protein binding and membrane association. This gymnotic delivery mechanism allows chemically modified oligonucleotides to enter neurons without transfection reagents, preserving cellular integrity. Following endocytic internalization, a productive fraction of ASOs escape endosomes to reach the cytosol and nucleus where they engage complementary mRNA targets and recruit RNase H1 for catalytic degradation. This approach extends to human iPSC-derived neurons for disease modeling applications, with uptake efficiency enhanced by calcium-enriched culture conditions . The transfection-free mechanism preserves neuronal morphology, electrophysiology, and synaptic function while achieving target-dependent knockdown in primary neurons (typical range 50-85% for most genes, with optimization required for each target).
Why Conventional Neuronal Transfection Methods Fail
Post-Mitotic Nature & Transfection Incompatibility
Neurons cease cell division after differentiation, creating fundamental incompatibility with conventional transfection. Advanced electroporation can directly deliver nucleic acids to the cytoplasm and nucleus of non-dividing neurons. Primary neurons are challenging for transfection due to their sensitivity to membrane disruption and selective uptake mechanisms, and while mitosis is not a requirement for ASO or siRNA delivery, post-mitotic neurons present unique challenges for conventional transfection methods. Consequently, lipofection efficiency in DIV 7+ neurons often drops below 5% , and electroporation can cause 50-80% immediate cell death due to membrane damage in non-dividing cells, though optimized protocols can preserve high viability . This makes functional studies in mature, physiologically relevant neurons challenging with conventional methods.
High ImpactElectrophysiological Disruption
Transfection reagents can alter neuronal membrane properties and ion channel function. Electroporation creates transient membrane pores that typically reseal within minutes to hours , though optimized protocols can preserve normal membrane properties. However, suboptimal electroporation conditions may temporarily affect input resistance and capacitance. Cationic lipids at high concentrations have been reported to affect membrane potential in some studies. These potential artifacts require careful optimization and controls for electrophysiology experiments studying neuronal excitability, synaptic transmission, and action potential propagation.
High ImpactNeurite Retraction and Morphological Damage
High-intensity transfection methods, particularly biolistic gene transfer and suboptimal electroporation, can cause morphological damage to neurons. Studies show that biolistic transfection with shRNA caused progressive dendritic spine loss (47% at 4 days, 54% at 7 days, and 88% at 14 days) in hippocampal neurons . Electroporation can reduce neurite outgrowth by approximately 44% at 48 hours in some neuronal subtypes . Neurons may retract neurite arbors under stress conditions, severing synaptic connections and disrupting neuronal network architecture. These morphological changes make it challenging to study neurite outgrowth, axon guidance, synaptogenesis, or circuit-level phenomena with certain transfection methods.
High ImpactViral Vector Toxicity in Long-Term Cultures
Lentiviral vectors can trigger interferon signaling and innate immune activation that alters baseline gene expression, though AAV vectors show minimal immunogenicity . However, insertional mutagenesis from lentiviral integration disrupts endogenous gene regulation, creating experimental artifacts. shRNA overexpression can saturate the Exportin-5 export pathway and cause widespread off-target effects by competing with endogenous miRNAs . Moreover, the 2-4 week timeline for viral production and validation is incompatible with primary neuronal culture windows (typically 3-4 weeks total).
Medium ImpactDevelopmental Stage Sensitivity
Neuronal transfection efficiency varies dramatically with developmental stage, creating a narrow and restrictive experimental window. Immature neurons (DIV 0-3) often lack fully developed uptake machinery and exhibit poor transfection tolerance, with viability frequently dropping to 40-50%. Mature neurons (DIV 7-21) develop resistance to transfection as they become fully post-mitotic, with lipofection efficiency often very low at DIV7-21 without specialized protocols. The narrow optimal window (DIV 3-5) restricts experiments to semi-mature neurons that may not fully recapitulate adult neuronal biology, limiting translational relevance.
Medium ImpactNeuronal Subtype Variability
Different neuronal subtypes exhibit wildly variable transfection susceptibility, requiring extensive cell-type-specific optimization. GABAergic interneurons show marginally higher transfection tolerance than glutamatergic pyramidal neurons, but still achieve only 10-20% efficiency with severe toxicity. Motor neurons are extremely fragile, with lipofection success rates below 10% and electroporation causing >80% death. DRG sensory neurons possess large cell bodies (30-50 μm diameter) that are highly resistant to transfection, requiring prohibitively high voltages that cause immediate cell lysis. Each neuronal subtype demands different protocols, making comparative studies across cell types impractical.
Medium ImpactMethod Comparison
| Method | Efficiency | Viability | Pros | Cons |
|---|---|---|---|---|
| Lipofection (Cationic Lipid Reagents) | 5-15% | 30-50% | Simple protocol, commercially available | Severe neurotoxicity, neurite retraction, disrupts electrophysiology, <5% in mature neurons (DIV 7+) |
| Electroporation Systems | 20-40% | 20-50% (standard protocols), 70-90% (optimized systems like Nucleofector™ with neuron-specific buffers) | Higher efficiency than lipofection | Severe cell death in post-mitotic neurons, irreversible morphological damage, alters membrane properties, expensive |
| Viral Vectors (Lentivirus, AAV) | 60-80% | 70-85% | High efficiency, long-term expression | Insertional mutagenesis, chronic inflammatory stress, 2-4 week production, safety concerns, off-target shRNA effects |
| AUMsilence sdASO | 70-95% | >90% | No transfection, preserves electrophysiology & morphology, works in post-mitotic neurons, no equipment needed | Transient knockdown (appropriate for acute functional studies) |
Why This Product?
Ideal For
- Primary rodent neurons (cortical, hippocampal, motor, DRG)
- Human iPSC-derived neurons (cortical, dopaminergic, motor)
- Post-mitotic mature neurons (DIV 7-21)
- Neurodegenerative disease modeling (AD, PD, ALS, HD)
- Synaptic plasticity and electrophysiology studies
- Pain research in DRG sensory neurons
- Neuronal cell lines (SH-SY5Y, PC12, N2a)
- Long-term neuronal cultures requiring preserved function
Alternative Products
When to use:
Learn More →When to use:
Learn More →AUMsilence Protocols for Primary Neurons
Cell-type-optimized protocols for cortical, hippocampal, motor, and sensory neurons. No transfection reagents required: preserves neuronal morphology and electrophysiology.
Quick Start Protocol (All Primary Neurons)
- Culture neurons in neuronal culture medium + supplement on poly-D-lysine/laminin-coated plates
- Add AUMsilence sdASO directly to culture medium at 10 μM (typical effective range: 5-20 μM depending on target and cell type; no transfection reagent )
- Incubate 48-72 hours at 37°C, 5% CO₂ (no media change required)
- Validate knockdown by qRT-PCR (mRNA), immunocytochemistry or Western blot (protein), and patch clamp electrophysiology (functional validation)
Cell-Type-Specific Protocols
Essential Controls for Neuronal RNA Silencing
Optimization Guide for Challenging Neuronal Applications
ASO Concentration
Recommendation: Start with 10 μM for most neuronal subtypes. Motor neurons and DRG neurons may require lower concentrations (5-7 μM) due to sensitivity. Highly stable transcripts may require 15-20 μM.
Rationale: Neuronal sensitivity varies by subtype. Lower concentrations reduce potential toxicity while higher concentrations overcome mRNA stability barriers.
Incubation Time
Recommendation: Standard: 48-72 hours. For very stable targets (structural proteins, ion channels): extend to 96-120 hours. For rapid-turnover targets (immediate early genes): 24-48 hours sufficient.
Rationale: Protein half-life determines time needed for phenotypic knockdown. Long-lived proteins require extended incubation for turnover.
Neuronal Culture Density
Recommendation: Cortical: 50,000-100,000 cells/cm². Hippocampal: 30,000-80,000 cells/cm². Motor neurons: 20,000-50,000 cells/cm². Optimize for robust synapse formation without excitotoxicity.
Rationale: Density affects network activity, glutamate accumulation, and ASO uptake. Too dense = excitotoxicity. Too sparse = poor synaptogenesis.
DIV Timing
Recommendation: Add ASO at DIV 7-14 for mature neurons. Earlier (DIV 3-5) for developmental studies. Avoid DIV 0-2 (immature, low uptake).
Rationale: Mature neurons (DIV 7+) have developed endocytic machinery and stable gene expression patterns. Immature neurons show variable ASO uptake.
Media Composition
Recommendation: Use neuronal culture medium with 2% B-27 or N2 supplement. Avoid serum (can bind ASOs). Include glutamine (2 mM) and glutamate (0.5 mM, first 24h only).
Rationale: Serum proteins can sequester ASOs, reducing effective concentration. Glutamate supports early neuronal survival but becomes excitotoxic if maintained long-term.
Co-Culture Considerations
Recommendation: For neuron-glia co-cultures: ASOs will enter both cell types. Use cell-type-specific markers to distinguish knockdown effects. Consider separate treatment if targeting glia-specific genes.
Rationale: Self-delivering ASOs enter all cell types in culture. Cell-type specificity comes from target gene expression pattern, not ASO delivery selectivity.
Validation Methods for Neuronal Knockdown
Comprehensive validation ensures robust gene silencing while preserving neuronal function. AUMsilence maintains >90% viability for all downstream assays.
Critical Controls for Validation
Untreated Neurons
Purpose: Baseline expression, viability, and electrophysiological properties
Culture neurons identically but without ASO addition. Use for all comparisons (qPCR, Western blot, patch clamp).
Non-Targeting Control ASO
Purpose: Control for off-target effects, immune activation, and ASO-related toxicity
Use AUM non-targeting control ASO at same concentration (standard 10 μM, typical effective range: 5-20 μM depending on target and cell type) and timing as experimental ASO. Should show no knockdown of target gene, normal viability, and normal electrophysiology.
Positive Control ASO (Housekeeping Gene)
Purpose: Verify ASO uptake and RNase H activity in neurons
Optional but recommended: Use GAPDH or ACTB-targeting ASO to confirm typical 70-95% knockdown. Validates that neurons are competent for ASO uptake.
Viability Control (Dead Neurons)
Purpose: Validate viability assay sensitivity
Induce neuronal death with glutamate (100 μM, 30 min) or hydrogen peroxide (200 μM, 1h) as positive control for cell death assays. Should show >80% death.
Electroporation/Lipofection Comparison
Purpose: Demonstrate superiority of AUMsilence over conventional transfection
Transfect parallel cultures with cationic lipid reagents or electroporation systems. Compare viability, morphology, and electrophysiology. Should show severe toxicity and neurite retraction with conventional methods.
Time Course Control
Purpose: Determine optimal validation timepoint for target gene
Harvest neurons at 24h, 48h, 72h, 96h post-treatment. Perform qPCR and Western blot to define mRNA and protein knockdown kinetics. Accounts for protein half-life variability.
Dose-Response Control
Purpose: Confirm concentration-dependent knockdown and identify optimal dose
Test at minimum 3 concentrations (e.g., 5 μM, 10 μM, 20 μM AUMsilence). Knockdown should correlate with concentration. Typical effective range: 5-20 μM depending on target and cell type. Plot dose-response curve to identify EC50.
Independent ASO Sequence Verification
Purpose: Confirm on-target specificity with second independent ASO
Design and test 3-5 different ASOs targeting non-overlapping regions of same mRNA. Concordant knockdown and phenotype across independent ASOs confirms on-target specificity and rules out off-target effects. This is gold standard for definitive target validation in neuroscience.
Best Practices
- SAFETY NOTE: Always verify target gene function before knockdown to avoid unexpected phenotypes. Some genes are essential for neuronal survival or have unknown roles in neuronal maintenance
- Use biological triplicates (n=3 independent neuronal cultures, different dissection dates) for statistical analysis
- Validate knockdown at both mRNA (qRT-PCR, 48-72h) and protein (Western/ICC, 72-96h) levels
- Perform electrophysiology (patch clamp or MEA) to confirm neuronal function preserved: this is the definitive validation for AUMsilence in neurons
- Include morphological analysis (neurite length, spine density) to demonstrate lack of neurotoxicity
- Use non-targeting control ASO at same concentration to rule out off-target effects
- For functional assays (LTP, calcium imaging), verify knockdown in same neurons used for functional readout
- Report both knockdown efficiency and cell viability in all publications and presentations
- Include electroporation or lipofection comparison to demonstrate AUMsilence advantage
- For ion channel targets, correlate mRNA/protein knockdown with functional current reduction (e.g., 80% Nav1.7 mRNA knockdown should produce 60-80% sodium current reduction)
- Use appropriate statistical tests: t-test for 2-group comparisons, one-way ANOVA with post-hoc for multiple groups, two-way ANOVA for interaction effects (e.g., genotype × treatment)
- Archive representative electrophysiology traces, calcium imaging movies, and microscopy images for supplementary data
Frequently Asked Questions
Ready to Advance Your Neuroscience Research?
Get expert guidance on designing transfection-free gene knockdown experiments in primary neurons. Our neuroscience team has extensive experience with electrophysiology, synaptic plasticity, and neurodegeneration models.
