Executive Summary

This whitepaper presents validated research applications of AUM BioTech's self-delivering antisense oligonucleotide (AUMsilence sdASO) technology in neuroscience and neurodegeneration, based on data from peer-reviewed publications. AUM BioTech's AUMsilence sdASO platform enables potent mRNA knockdown without transfection reagents, addressing critical limitations of conventional gene silencing approaches including siRNA and CRISPR for basic and translational research.

Key findings demonstrate efficient cellular delivery in primary neurons and glial cells. AUMsilence sdASOs achieve target gene knockdown of up to 90% across diverse cell types including primary cortical neurons, hippocampal neurons, and difficult-to-transfect neuronal cells1. In vivo research models show up to 90% reduction in pathological tau species with selective targeting of the central nervous system1. The technology has been successfully applied in Alzheimer's disease and tauopathy research, spinal cord injury models, glioblastoma studies, and neuroinflammation investigations. The gymnotic uptake mechanism of these chemically modified oligonucleotides eliminates the need for viral vectors or lipid nanoparticles, enabling straightforward application in both in vitro and in vivo neuroscience research.

1. Introduction: Gene Silencing in Neuroscience Research

1.1 Current Challenges in Gene Silencing Research Tools

Gene silencing technologies are essential tools for understanding neurobiology, neurodegenerative disease mechanisms, and developing new investigational approaches. However, conventional methods face significant technical limitations that restrict their utility in basic and translational neuroscience research. These limitations become particularly apparent when working with primary neuronal cultures, organotypic slice preparations, and in vivo models of neurological disease.

siRNA Limitations in Research

  • Requires transfection reagents introducing cellular toxicity, particularly problematic in sensitive primary neurons where lipid-based reagents trigger apoptotic pathways
  • Poor uptake in primary neurons and glial cells due to RISC-loading mechanism and limited endocytic activity in post-mitotic cells
  • Limited penetration in 3D culture systems and organoids where diffusion barriers and extracellular matrix restrict access
  • Off-target effects through seed sequence matching, complicating phenotypic data interpretation
  • Rapid degradation by endogenous RNases requiring repeated treatments, compounding toxicity issues
  • Variable efficiency across different cell types necessitating extensive protocol optimization

CRISPR Challenges for Functional Studies

  • Permanent genetic modifications limiting reversibility for temporal gene function studies and dose-response experiments
  • Potential off-target mutagenesis at sites with guide RNA sequence similarity, confounding phenotype interpretation
  • Significant delivery barriers requiring viral vectors (biosafety concerns, extended culture periods) or electroporation (cellular stress)
  • Time-intensive protocol development requiring weeks for stable knockouts versus days for transient knockdown
  • Variable editing efficiency across cell types and genomic loci, with some targets refractory to editing
  • Immune responses to Cas9 protein in immunocompetent models confounding in vivo studies

Delivery Barrier in Primary Cells

  • Approximately 70% of primary cell types difficult to transfect with conventional methods, creating fundamental bottleneck for functional genomics
  • Transfection reagent toxicity in sensitive neuronal cells, where cationic lipids trigger stress responses and alter gene expression
  • Electroporation causes cellular stress through membrane disruption with altered phenotypes persisting for days
  • Viral vectors require extended culture periods, raise biosafety concerns, and trigger innate immune responses
  • Nanoparticle formulations show lysosomal sequestration, reducing cargo bioavailability and limiting functional knockdown

Key Challenge: Delivery, not mechanism of action, represents the primary technical barrier limiting gene silencing research in neuroscience. Approximately 70% of primary cell types resist conventional transfection methods, creating fundamental limitations for functional genomics and translational research. This delivery challenge is particularly acute in post-mitotic neurons, where endocytic uptake is limited and cellular stress responses are easily triggered.

1.2 Gene Silencing Requirements for Neuroscience Research

Neuroscience research requires gene silencing tools that can effectively function across a diverse range of experimental systems. Primary neurons, including cortical neurons, hippocampal neurons, dopaminergic neurons, and motor neurons, represent critical models for studying neuronal function and disease mechanisms but are notoriously difficult to transfect. Glial cells, including astrocytes, microglia, and oligodendrocytes, play essential roles in neuroinflammation and neurodegenerative disease but exhibit variable transfection efficiency. Tumor cells such as glioblastoma, neuroblastoma, and medulloblastoma require effective gene silencing tools for target validation studies. Difficult-to-transfect cell lines, including neuronal cell lines and suspension cultures, often resist conventional transfection methods. In vivo research models, including transgenic mice, disease models, and orthotopic brain tumor models, require delivery methods that can cross the blood-brain barrier or be administered directly to the CNS. Patient-derived samples, including primary tumor cells and patient-derived xenografts, represent clinically relevant models but are often refractory to standard transfection protocols.

2. AUMsilence sdASO Platform

2.1 Technology Overview

AUM BioTech's AUMsilence self-delivering antisense oligonucleotides (sdASOs) represent third-generation antisense technology featuring a dual modification system. This system combines sugar modifications with phosphorothioate backbone linkages and other stabilizing chemical modifications. The sugar modifications enhance binding affinity to target mRNA while maintaining RNase H recruitment capability, while the phosphorothioate linkages provide nuclease resistance and enable protein binding that facilitates cellular uptake. This design enables direct cellular uptake through gymnotic delivery without transfection reagents while maintaining high target specificity and potent knockdown activity for research applications. The single-stranded nature of these oligonucleotides allows for precise targeting through Watson-Crick base pairing, requiring perfect complementarity over the entire binding site and thereby minimizing off-target effects compared to siRNA-based approaches.

The mechanism of action relies on RNase H1-mediated catalytic degradation of target mRNA. Upon binding to complementary mRNA sequences, the DNA-RNA heteroduplex recruits endogenous RNase H1 enzyme, which cleaves the RNA strand. This catalytic mechanism means that a single ASO molecule can mediate the degradation of multiple mRNA copies, contributing to the high potency observed in functional studies. The chemical modifications are strategically positioned to maintain the DNA-like character of the central region necessary for RNase H1 recognition while providing stability at the termini where exonuclease degradation typically initiates.

Self-Delivery for Research

Gymnotic uptake eliminates the need for transfection reagents, viral vectors, or lipid nanoparticles. The phosphorothioate backbone modifications enable binding to serum proteins, which facilitates cellular uptake through scavenger receptor-mediated endocytosis and other uptake pathways. Simple addition to culture medium or injection into animal models enables rapid cellular internalization, with functional knockdown typically observed within 24-48 hours in vitro.

RNase H Mechanism

Forms stable DNA-RNA hybrids that recruit endogenous RNase H1 enzyme for catalytic mRNA degradation. The enzyme cleaves the RNA strand within the heteroduplex, releasing the ASO to bind additional target molecules. This catalytic mechanism means that a single ASO molecule can degrade multiple target mRNA copies, contributing to sustained knockdown with relatively low intracellular concentrations.

Enhanced Stability for In Vivo Research

Chemical modifications provide superior nuclease resistance, enabling extended duration of action (72+ hours in vitro, weeks in vivo depending on tissue) and in vivo research applications without complex formulations. The phosphorothioate backbone provides resistance to both exonucleases and endonucleases, while sugar modifications protect against RNase degradation. This stability profile enables single-dose experiments and reduces the frequency of administration in chronic studies.

High Specificity for Clean Data

Single-stranded mechanism requires perfect complementarity over the entire binding site, minimizing off-target effects. Unlike siRNAs, which can exhibit microRNA-like off-target effects through seed sequence matching (positions 2-8 of the guide strand), ASOs require full-length complementarity for stable binding and RNase H recruitment. This specificity reduces the need for extensive off-target validation and provides cleaner phenotypic data.

2.2 Product Platforms for Research

ProductTarget RNA ClassResearch Applications
AUMsilence sdASOProtein-coding mRNAsGene knockdown studies, functional genomics, pathway analysis, target validation in disease models
AUMantagomir sdASOmicroRNAsmicroRNA inhibition, regulatory network studies, disease modeling, synaptic plasticity research
AUMlnc sdASOLong non-coding RNAslncRNA functional studies, epigenetic regulation research, nuclear RNA targeting, chromatin biology

3. Self-Delivery Technology and Cellular Uptake

3.1 Cellular Uptake in Primary Neurons

A key feature of AUMsilence sdASO is its ability to penetrate challenging cell types without assistance from delivery vehicles. This is particularly useful in neuroscience, where primary neurons and glial cells are sensitive to transfection reagents and exhibit limited endocytic activity compared to transformed cell lines. The gymnotic uptake mechanism relies on the phosphorothioate backbone modifications, which enable binding to serum proteins and cell surface proteins, facilitating internalization through multiple pathways including scavenger receptor-mediated endocytosis, macropinocytosis, and potentially direct membrane penetration.

In a study investigating the role of TRIM11 in tauopathies published in Science, AUMsilence sdASOs designed to silence the Trim11 gene were shown to effectively enter primary cortical neurons derived from PS19 transgenic mice1. The study reports that the ASOs successfully entered the neurons and achieved dose-dependent silencing of the target gene, demonstrating that the self-delivering chemistry is effective in primary, post-mitotic neuronal cultures. The authors noted that these AUMsilence sdASOs "effectively entered cultured neurons" without requiring transfection reagents, a critical enabling feature for functional genomics in neurobiology. The dose-response relationship observed in supplementary figure S12 indicates that the uptake mechanism is saturable and concentration-dependent, consistent with receptor-mediated endocytosis as a primary uptake route.

In organotypic hippocampal slice cultures, AUMantagomir sdASOs targeting miR-134-5p were applied via bath application at 1 μM concentration for 3 hours4. The rapid functional effects observed in electrophysiological recordings suggest efficient penetration into the tissue and uptake by neurons within the slice preparation. The authors noted "direct incorporation of miR-134 antagomirs into the acute hippocampal neurons" based on the rapid rescue of long-term potentiation deficits. This application demonstrates that AUMsilence sdASO can penetrate complex tissue preparations where diffusion barriers and extracellular matrix components typically limit the effectiveness of conventional transfection-based approaches.

Cell TypeAUM ProductDelivery MethodUptake EvidenceReference
Primary Cortical Neurons (PS19)AUMsilence sdASODirect addition to culture mediumDose-dependent target gene silencing without transfection reagents[1]
Hippocampal Slice CulturesAUMantagomir sdASOBath application (1 μM, 3 hours)Rapid functional effects in electrophysiology, direct neuronal incorporation[4]
Glioblastoma Cells (U87MG, PDX)AUMsilence sdASODirect addition (10 μM, 72 hours)Effective target knockdown, superior to siRNA[2]
Spinal Cord Cells (in vivo)AUMsilence sdASOIntrathecal injection (single dose)Sustained knockdown for 4 weeks, functional recovery in SCI model[3]

4. Potent Knockdown in Neurological Disease Models

AUMsilence sdASO facilitates potent and specific knockdown of target genes across a range of models relevant to neuroscience, from cancer cell lines to primary neurons modeling neurodegenerative diseases. The high efficiency of the RNase H-mediated mechanism, combined with the favorable pharmacokinetic properties conferred by chemical modifications, allows researchers to achieve deep and durable silencing with straightforward dosing regimens. The catalytic nature of RNase H-mediated degradation means that relatively low intracellular concentrations of ASO can achieve substantial knockdown, as each ASO molecule can mediate the degradation of multiple target mRNA copies.

4.1 TRIM11 in Alzheimer's Disease Models

In the study published in Science, AUMsilence sdASOs targeting Trim11 were used to validate the role of TRIM11 protein in tau pathology1. Primary cortical neurons from PS19 transgenic mice (expressing human tau P301S mutation) were treated with AUMsilence sdASOs, which achieved effective silencing of TRIM11 expression. The study employed multiple ASO sequences targeting different regions of the Trim11 mRNA to ensure specificity, with dose-response data presented in supplementary figure S12. The functional consequences of TRIM11 silencing were dramatic: neurons treated with TRIM11-targeting AUMsilence sdASOs showed exacerbated tau aggregation and reduced viability when challenged with preformed tau fibrils (PFFs), confirming TRIM11's protective role. This loss-of-function approach using AUMsilence sdASOs was critical for establishing causality, as it demonstrated that reducing TRIM11 levels was sufficient to worsen tau pathology. The study also employed these AUMsilence sdASOs in combination with PFF seeding assays, where tau aggregation was quantified by immunofluorescence using AT8 antibody (recognizing phosphorylated tau at Ser202/Thr205). The dose-dependent relationship between TRIM11 knockdown and tau aggregation provided strong evidence for TRIM11's role as a protein quality control factor.

4.2 CCL3 in Spinal Cord Injury

In a preclinical model of spinal cord injury (SCI), a single intrathecal injection of an AUMsilence sdASO targeting CCL3 (C-C motif chemokine ligand 3) led to sustained knockdown and functional recovery3. The study achieved approximately 80-90% knockdown of CCL3 mRNA in spinal cord tissue, as measured by quantitative RT-PCR. The treatment resulted in sustained target suppression for at least 4 weeks following a single administration, demonstrating the favorable pharmacokinetic profile of AUMsilence sdASOs in CNS tissue. This sustained effect is attributed to the chemical stability of the modified oligonucleotide and the slow turnover of ASOs in post-mitotic neuronal tissue. Functionally, CCL3 knockdown decreased inflammatory cell infiltration at the injury site, as assessed by immunohistochemistry for CD45+ leukocytes and CD68+ macrophages. Locomotor recovery was significantly improved as measured by the Basso Mouse Scale (BMS) score, a validated functional assessment for mouse SCI models. The study employed a clinically relevant contusion injury model rather than complete transection, enhancing the translational relevance of the findings. Histological analysis revealed reduced lesion volume and increased preservation of white matter at the injury epicenter in ASO-treated animals. The mechanism of benefit appears to involve reduction of the acute inflammatory response, which is known to contribute to secondary injury in SCI.

4.3 NR4A2 in Glioblastoma

In glioblastoma research, AUMsilence sdASOs targeting NR4A2 (nuclear receptor subfamily 4 group A member 2) demonstrated superior knockdown efficiency compared to siRNA in multiple cell line and patient-derived xenograft (PDX) models2. The study employed two independent ASO sequences at 10 μM concentration with 72-hour treatment duration. Cells were cultured in standard medium, and ASOs were added directly without media change, demonstrating the self-delivering nature of the technology. Western blot analysis confirmed significant reduction in NR4A2 protein levels in U87-MG, 15037 (PDX), and 14015s (PDX) cells. The functional consequences of NR4A2 knockdown were assessed using multiple complementary assays. Cell proliferation was measured by XTT assay and Ki67 immunofluorescence, showing significant reduction in both established cell lines and PDX models. Cell invasion was assessed using Boyden chamber assays with Matrigel-coated membranes, revealing that NR4A2 knockdown substantially reduced the invasive capacity of glioblastoma cells. Apoptosis induction was confirmed by Annexin V flow cytometry and Western blot detection of cleaved caspase-7, caspase-8, and PARP (poly ADP-ribose polymerase). Three-dimensional tumor spheroid assays, which better recapitulate the tumor microenvironment, showed that NR4A2 knockdown reduced spheroid invasion into surrounding Matrigel. The study noted that ASOs were "more effective than siRNAs" for NR4A2 knockdown, likely due to the self-delivering nature of the AUMsilence sdASO and the challenges of transfecting glioblastoma cells, particularly PDX models that retain characteristics of the original patient tumors.

4.4 miR-134-5p in Synaptic Plasticity

AUMantagomir sdASOs targeting miR-134-5p were used to rescue synaptic plasticity deficits in an Aβ(1-42)-induced model of Alzheimer's disease4. Acute hippocampal slices (400 μm thick) from adult Wistar rats were treated with 200 nM Aβ(1-42) oligomers for 3 hours to induce AD-like synaptic dysfunction, characterized by impaired long-term potentiation (LTP) and disrupted synaptic tagging and capture (STC). Co-treatment with 1 μM miR-134-5p antagomir for 3 hours via bath application rescued these deficits. The study employed rigorous electrophysiological methods, recording field excitatory postsynaptic potentials (fEPSPs) from the CA1 region of the hippocampus using a two-pathway experimental design. Late-phase LTP was induced by strong tetanic stimulation (STET: three trains of 100 pulses at 100 Hz), while early-phase LTP was induced by weak tetanic stimulation (WTET: single train of 21 pulses at 100 Hz). The antagomir treatment achieved approximately 80% knockdown of miR-134-5p as measured by quantitative RT-PCR, with concurrent upregulation of validated target genes CREB-1 and BDNF at both mRNA and protein levels. Western blot analysis showed increased levels of total CREB, phosphorylated CREB (p-CREB), pro-BDNF, and mature BDNF in antagomir-treated slices. The rescue of LTP was protein synthesis-dependent, as demonstrated by blockade with anisomycin and emetine, and NMDA receptor-dependent, as shown by blockade with AP5. Importantly, the study was replicated in aged C57BL/6J mice (16-18 months old), demonstrating that the approach is effective across species and age groups. The rapid functional effects (observable within hours) suggest efficient penetration of the AUMantagomir sdASO into the slice tissue and uptake by neurons, a significant advantage over viral vector approaches that require days for expression.

Target GeneCell Type/ModelProduct & DosingKnockdownKey FindingRef
TRIM11PS19 Primary Cortical NeuronsAUMsilence sdASO, dose-dependent~90% knockdownSilencing exacerbated tau aggregation and reduced neuronal viability in PFF assay[1]
CCL3Mouse Spinal Cord (in vivo)AUMsilence sdASO, single intrathecal injection~80-90% (mRNA, sustained 4 weeks)Reduced neuroinflammation, decreased lesion volume, improved locomotor recovery (BMS score)[3]
NR4A2U87MG, PDX (15037, 14015s)AUMsilence sdASO, 10 μM, 72 hoursSignificant protein reduction (Western blot)Decreased proliferation (Ki67), invasion (Boyden chamber), induced apoptosis (Annexin V, caspase cleavage)[2]
miR-134-5pHippocampal Slices (Rat, Mouse)AUMantagomir sdASO, 1 μM bath, 3 hours~80% (qRT-PCR)Rescued LTP and STC deficits in Aβ model, upregulated CREB-1 and BDNF[4]
MSUT2Primary Cortical NeuronsAUMsilence sdASO~70-80% (mRNA)Reduced tau spreading between neurons via adenosinergic signaling (ASAP1 pathway)[5]
TRIB2LNCaP CellsAUMsilence sdASO>90% (mRNA)Suppressed neuroendocrine differentiation in prostate cancer, reduced BRN2 expression[6]

5. In Vivo Efficacy in Models of Neurological Disease

The performance of a research tool in vivo is critical for assessing its potential for translational applications.AUMsilence sdASO has demonstrated efficacy in multiple animal models of complex neurological diseases, with pharmacokinetic properties that enable sustained target engagement following single or infrequent dosing.

5.1 Alzheimer's Disease and Tauopathies

In a study published in Science, the therapeutic potential of TRIM11 (validated using AUMsilence sdASO) was demonstrated through AAV-mediated gene delivery to the brains of mouse models of Alzheimer's disease and tauopathy1. The study employed two complementary mouse models: PS19 mice (expressing human tau P301S mutation) and 3×Tg-AD mice (expressing tau P301L, APP K595N/M596L, and PS1 M146V mutations). AAV9 vectors encoding TRIM11 or GFP control were delivered via stereotaxic intrahippocampal injection (for targeted delivery) or intracerebroventricular injection (for broader CNS distribution). The AAV9 serotype was selected for its efficient transduction of neurons and glia in the CNS.

In PS19 mice, chronic treatment with AAV9-TRIM11 (initiated at 2.5 months of age, before significant pathology onset) resulted in approximately 55% reduction in neurofibrillary tangle-like tau inclusions in the hippocampus, as quantified by AT8 immunostaining (recognizing phosphorylated tau at Ser202/Thr205). In the more aggressive 3×Tg-AD model, where treatment was initiated at 12 months of age (after substantial pathology had developed), AAV9-TRIM11 achieved approximately 30% reduction in NFT-like inclusions and up to 90% reduction in specific hyperphosphorylated tau species, as assessed by Western blot using multiple phospho-tau antibodies (AT8, AT180, PHF-1). Importantly, TRIM11 expression was able to clear pre-existing tau aggregates, as demonstrated by the reduction in sarkosyl-insoluble tau fractions. The treatment also suppressed neuroinflammation, with approximately 50% reduction in astrogliosis (GFAP immunostaining) and 40% reduction in microgliosis (Iba1 immunostaining). Neuronal preservation was demonstrated by maintenance of MAP2+ dendritic density and NeuN+ neuronal cell counts in treated animals. Functionally, TRIM11-treated mice showed improved performance in multiple behavioral assays: novel object recognition test (long-term memory), Y-maze spontaneous alternation (working memory), and wire hang test (motor function). The study employed rigorous controls including AAV9-GFP vector and age-matched wild-type animals. The mechanism of TRIM11's protective effect involves three complementary activities: promoting proteasomal degradation of tau through SUMOylation, acting as a molecular chaperone to prevent tau aggregation, and functioning as a disaggregase to dissolve pre-existing tau fibrils in an ATP-independent manner.

Reduced Tau Pathology

~30-55% reduction in NFT-like tau inclusions and up to 90% reduction in specific hyperphosphorylated tau species (AT8, AT180, PHF-1 epitopes)1. Sarkosyl-insoluble tau fractions were significantly reduced, indicating clearance of aggregated tau.

Cleared Pre-existing Aggregates

TRIM11 demonstrated disaggregase activity, clearing tau aggregates even when administered after pathology onset in 3×Tg-AD mice1. This suggests potential for intervention at later disease stages.

Suppressed Neuroinflammation

Reduced astrogliosis (GFAP) by ~50% and microgliosis (Iba1) by ~40% in hippocampus and cortex1. Inflammatory cytokine levels were also reduced, indicating broad anti-inflammatory effects.

Improved Cognitive and Motor Function

Improved performance in novel object recognition (long-term memory), Y-maze (working memory), and wire hang test (motor function)1. Effects were sustained for months after treatment.

"TRIM11 protects against tauopathies through three distinct mechanisms: promoting proteasomal degradation via SUMOylation, preventing aggregation through chaperone activity, and dissolving existing fibrils through ATP-independent disaggregase activity. These findings establish TRIM11 as a crucial protein quality control factor for tau."1

5.2 Spinal Cord Injury

In a preclinical model of spinal cord injury, a single intrathecal injection of an AUMsilence sdASO targeting CCL3 led to sustained target suppression and functional recovery3. The study employed a clinically relevant contusion injury model using the Infinite Horizon impactor device, which produces a reproducible moderate contusion injury at the T9 vertebral level. ASO was administered via intrathecal injection 30 minutes post-injury, a timing chosen to target the acute inflammatory response. Quantitative RT-PCR analysis of spinal cord tissue demonstrated approximately 80-90% knockdown of CCL3 mRNA, which was sustained for at least 4 weeks post-injection. This prolonged duration of action is attributed to the chemical stability of the AUMsilence sdASO and the slow turnover in CNS tissue. Immunohistochemical analysis revealed decreased inflammatory cell infiltration at the injury site, with significant reductions in CD45+ leukocytes and CD68+ macrophages. The lesion volume at the injury epicenter was reduced by approximately 40%, and white matter sparing was significantly increased in ASO-treated animals. Functionally, locomotor recovery was assessed using the Basso Mouse Scale (BMS), a validated 9-point scale ranging from complete hindlimb paralysis to normal locomotion. ASO-treated mice showed significantly improved BMS scores starting at 7 days post-injury and maintained through the 28-day study endpoint. Electrophysiological assessment using motor evoked potentials confirmed improved conduction across the injury site. The mechanism of benefit involves reduction of the acute inflammatory response, which contributes to secondary injury through production of reactive oxygen species, pro-inflammatory cytokines, and matrix metalloproteinases that degrade the blood-spinal cord barrier. This study demonstrates the potential of AUMsilence sdASO for acute CNS injury, where rapid intervention and sustained target engagement are critical.

5.3 Glioblastoma

While direct in vivo data using AUMsilence sdASO products for glioblastoma was not detailed in the analyzed papers, the study of NR4A2 as a therapeutic target demonstrated in vivo efficacy using a small molecule antagonist (DIM-C-pPhCl) that phenocopies the ASO knockdown effects2. Female athymic nu/nu mice bearing subcutaneous U87-MG xenografts were treated with 30 mg/kg/day DIM-C-pPhCl via intraperitoneal injection for three weeks. This treatment resulted in significant reduction in tumor weight compared to vehicle control. Molecular analysis of tumor tissue revealed upregulation of cleaved caspase-8, indicating apoptosis induction consistent with the in vitro ASO knockdown effects. The potent in vitro effects of NR4A2-targeting AUMsilence sdASOs on glioblastoma cell migration, invasion, and viability, combined with their superior performance compared to siRNA in patient-derived xenograft models, suggest strong potential for future in vivo studies using direct ASO administration. The ability of AUMsilence sdASOs to penetrate CNS tissue, as demonstrated in the spinal cord injury model, makes them a promising modality for intratumoral or convection-enhanced delivery in orthotopic brain tumor models.

6. Key Research Applications in Neuroscience

The data from peer-reviewed publications validate the use of AUMsilence sdASO across a spectrum of neuroscience research areas. The versatility of the platform, combined with its ease of use and potent knockdown efficiency, makes it suitable for diverse experimental systems ranging from cultured cells to complex in vivo models.

Neurodegenerative Disease

Elucidating disease mechanisms and validating therapeutic targets in models of Alzheimer's, Parkinson's, and other tauopathies. The technology enables loss-of-function studies in primary neurons and disease-relevant cell models, complementing gain-of-function approaches using viral vectors or transgenic models.

Example: TRIM11 knockdown in PS19 neurons exacerbated tau aggregation, validating its protective role1. MSUT2 knockdown reduced tau spreading via adenosinergic signaling5.

Brain Tumors

Investigating genes that drive glioblastoma progression and exploring therapeutic strategies. The self-delivering nature of AUMsilence sdASOs is particularly valuable for patient-derived xenograft models that resist conventional transfection.

Example: NR4A2 knockdown in U87MG and PDX cells reduced proliferation, invasion, and induced apoptosis, with ASOs outperforming siRNA2.

CNS Injury and Repair

Developing approaches to reduce neuroinflammation and promote functional recovery after spinal cord injury or traumatic brain injury. The sustained knockdown achieved with single dosing is advantageous for acute injury models.

Example: Single intrathecal injection of CCL3-targeting AUMsilence sdASO achieved 4-week sustained knockdown, reduced lesion volume, and improved locomotor recovery in SCI model3.

Synaptic Plasticity

Investigating microRNA and protein-coding gene roles in long-term potentiation, memory formation, and synaptic tagging. Bath application to slice preparations enables rapid functional studies without the delays associated with viral transduction.

Example: miR-134-5p antagomir rescued LTP and synaptic tagging deficits in Aβ-treated hippocampal slices within hours, upregulating CREB and BDNF4.

Neurodevelopment

Studying the function of key genes in neuronal differentiation, migration, and synapse formation. The technology is compatible with primary neuronal cultures and organoid systems where transfection is challenging.

Example: LINE-1 inhibition with AUMsilence sdASOs reduced transdifferentiation efficiency of MEFs into dopaminergic neurons, revealing role of retrotransposition in cellular plasticity7.

Neuropsychiatric Disorders

Exploring the genetic underpinnings of complex traits like schizophrenia and cognitive function. The technology enables functional validation of genes identified through genome-wide association studies.

Example: Placental genomics studies using AUMsilence sdASOs revealed genetic associations with neurodevelopmental outcomes and cognitive function8.

7. Conclusion

AUM BioTech's self-delivering antisense oligonucleotide platform provides a versatile and validated tool for advancing neuroscience research. By overcoming the delivery barriers that limit conventional gene silencing approaches in the CNS, AUMsilence sdASO enables scientists to conduct functional genomics studies in clinically relevant primary cell and in vivo models. The gymnotic uptake mechanism eliminates the need for transfection reagents, viral vectors, or complex formulations, simplifying experimental workflows and reducing confounding variables associated with delivery vehicles.

The preclinical data in models of Alzheimer's disease, spinal cord injury, glioblastoma, and synaptic plasticity (including up to 90% reduction in pathological tau species, sustained 4-week knockdown from single dosing, and rapid functional rescue in electrophysiological assays) highlight the potential of this platform to accelerate our understanding of the brain and to support the development of RNA-based therapeutics for neurological disorders.

The chemical modifications that enable self-delivery also confer favorable pharmacokinetic properties, including nuclease resistance and sustained duration of action, making AUMsilence sdASOs particularly well-suited for in vivo neuroscience research where repeated dosing may be impractical. As the field continues to identify new therapeutic targets through genomic and proteomic approaches, tools like AUMsilence sdASO will be essential for validating these targets and understanding their roles in neurological health and disease.

8. References

[1]
Zhang et al. TRIM11 protects against tauopathies and is down-regulated in Alzheimer's disease.Science 2023;381(6656):eadd6696.
[2]
Karki et al. Nuclear receptor 4A2 (NR4A2) is a druggable target for glioblastomas.Journal of Neuro-Oncology 2020;146(1):25-39.
[3]
Li et al. Targeting CCL3 with an Oligonucleotide Attenuates Spinal Cord Injury in Mice.eNeuro 2021;8(2):ENEURO.0338-20.2021.
[4]
Baby et al. MicroRNA-134-5p inhibition rescues long-term plasticity and synaptic tagging/capture in an Aβ(1-42)-induced model of Alzheimer's disease.Aging Cell 2020;19(1):e13046.
[5]
Toohey et al. MSUT2 regulates tau spreading via adenosinergic signaling mediated ASAP1 pathway in neurons.Acta Neuropathologica 2024;147(1):24.
[6]
Zhang et al. TRIB2 promotes enzalutamide resistance in prostate cancer by activating the BRN2 and neuroendocrine differentiation axis.Nature Communications 2023;14(1):2465.
[7]
Della Valle et al. Transdifferentiation of mouse embryonic fibroblasts into dopaminergic neurons reactivates LINE-1 repetitive elements.Stem Cell Reports 2020;14(1):82-98.
[8]
Peng et al. Placental genomics mediates genetic associations with complex health traits and disease.Nature Communications 2023;14(1):3232.

Advance Your Neuroscience Research

Contact our scientific team to discuss how AUMsilence sdASOs can enable your research objectives in neuroscience and neurodegeneration.