The C9orf21 protein, encoded by the C9ORF72 gene, is associated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) . A hexanucleotide repeat expansion (HRE) in an intron of the C9ORF72 gene is the most common cause of familial ALS and FTD . This expansion leads to decreased expression of the C9ORF72 protein and the production of toxic dipeptide repeat (DPR) proteins through repeat-associated non-ATG (RAN) translation .
The C9ORF72 gene expansion leads to several consequences:
Reduced C9ORF72 levels Decreased expression of the C9ORF72 protein impairs autophagy, hindering the clearance of DPR proteins .
DPR protein production Sense and antisense repeats are translated into toxic DPR proteins, including poly-GA, poly-PG, and poly-GR . Arginine-rich DPR proteins are particularly toxic to motor neurons, disrupting the ubiquitin-proteasome system (UPS) .
Autophagy impairment Reduced C9ORF72 levels result in suboptimal autophagy, leading to the accumulation of DPR proteins and neuronal cell death .
The C9ORF72 protein plays a significant role in neurodegenerative diseases, particularly ALS and FTD . The double-hit pathogenic mechanism involves reduced C9ORF72 expression and DPR protein accumulation, leading to neuronal cell death .
Several therapeutic strategies are being explored to target C9ORF72-related pathology:
20S Proteasome Enhancers Enhancing the degradation of DPR proteins using 20S proteasome activators like TCH-165 can protect motor neurons from DPR protein toxicity and restore proteostasis .
Targeting RAN protein production Inhibiting RAN protein production is explored as RAN proteins are considered toxic and unnecessary for normal cellular function1. PKR k296R, a mutant version of PKR protein, and metformin, a type 2 diabetes drug, can inhibit the pathway that upregulates the production of these proteins1.
Antibody Treatment Anti-GA antibodies have shown promise in reducing RAN proteins and improving motor neuron survival and longevity in mice1.
Autophagy Activation Pharmacological compounds that activate autophagy can prevent neuronal cell death caused by DPR protein accumulation .
C9ORF72 and its associated DPR proteins can impact multiple cellular pathways:
Ubiquitin-Proteasome System (UPS) Arginine-rich DPR proteins disrupt the UPS, affecting protein degradation .
Autophagy Reduced C9ORF72 levels impair autophagy, leading to the accumulation of toxic proteins .
Neuroinflammation Metformin has shown potential in decreasing neuroinflammation in mice with C9ORF72-related pathology1.
Genome-wide analysis reveals genes that are up-regulated and down-regulated under hypoxic conditions, offering insights into potential therapeutic targets.
| Gene | Description | Cobalt Treatment (C) | Hypoxia (H) |
|---|---|---|---|
| ALDOA | aldolase A, fructose-bisphosphate | 1.74 | -1.32 |
| ALDOC | aldolase C, fructose-bisphosphate | 6.93 | -3.57 |
| ANG | angiogenin, ribonuclease, RNase A family, 5 | 1.61 | -1.79 |
| ANKRD37 | ankyrin repeat domain 37 | 5.43 | -3.73 |
| HMOX1 | heme oxygenase (decycling) 1 | 6.47 | -2.87 |
| INSIG2 | insulin induced gene 2 | 2.57 | -2.11 |
| MAFF | v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (avian) | 1.87 | -1.48 |
| PDK1 | pyruvate dehydrogenase kinase, isozyme 1 | 2.56 | -1.18 |
| PFKFB3 | 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 | 1.55 | -1.41 |
| PGM1 | phosphoglucomutase 1 | 1.97 | -1.41 |
| SPAG4 | sperm associated antigen 4 | 2.88 | -1.19 |
| TMEM45A | transmembrane protein 45A | 5.45 | -2.53 |
| ZNF292 | zinc finger protein 292 | 2.69 | -1.56 |
| ABCB6 | ATP-binding cassette, sub-family B (MDR/TAP), member 6 | 2.73 | -1.90 |
| ANKZF1 | ankyrin repeat and zinc finger domain containing 1 | 2.74 | -1.75 |
| CITED2 | Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 2 | 1.71 | -1.87 |
| CSRP2 | cysteine and glycine-rich protein 2 | 2.10 | -2.05 |
| FOS | FBJ murine osteosarcoma viral oncogene homolog | 2.99 | -1.83 |
| GYS1 | glycogen synthase 1 (muscle) | 1.62 | -1.27 |
| HCFC1R1 | host cell factor C1 regulator 1 (XPO1 dependent) | 1.69 | -1.87 |
| KDM4B | lysine (K)-specific demethylase 4B | 2.21 | -1.82 |
| LIMCH1 | LIM and calponin homology domains 1 | 1.99 | -1.46 |
| RAB20 | RAB20, member RAS oncogene family | 3.88 | -2.20 |
| WSB1 | WD repeat and SOCS box-containing 1 | 1.79 | -1.29 |
| YEATS2 | YEATS domain containing 2 | 1.58 | -1.26 |
| SPOCK1 | sparc/osteonectin, cwcv and kazal-like domains proteoglycan (testican) 1 | 1.57 | -1.37 |
| EEF1E1 | eukaryotic translation elongation factor 1 epsilon 1 | -1.68 | -1.01 |
| BOP1 | block of proliferation 1 | -1.68 | 1.46 |
| DDX21 | DEAD (Asp-Glu-Ala-Asp) box polypeptide 21 | -1.61 | 1.56 |
| IL18R1 | interleukin 18 receptor 1 | -1.95 | 1.97 |
To expand your research, consider the following resources:
PubMed For additional studies on C9orf21 and related proteins, visit PubMed .
STRING Database Explore protein-protein interactions involving C9orf21 using the STRING database .
Google Patents Review relevant patents for potential therapeutic applications .
PMC Examine published articles related to C9orf21, ALS, and FTD in PMC .
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for fulfillment based on your requirements.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Tag type is determined during production. If you require a specific tag, please inform us for preferential development.
May positively regulate ERK1/2 signaling and AKT1 activation, leading to HIF1A upregulation, increased expression of glycolysis genes, and enhanced glycolysis.
UPF0308 protein C9orf21 is a 226 amino acid protein encoded by a gene mapped to human chromosome 9q22.33. It belongs to the UPF0308 protein family and is also known by several alternative designations:
AAED1 (AhpC/TSA antioxidant enzyme domain containing 1)
PRXL2C (Peroxiredoxin-like 2C)
Thioredoxin-like protein AAED1
The protein has subcellular localization in both the cytoplasm and nucleus. Chromosome 9, where C9orf21 is located, consists of approximately 145 million bases, representing 4% of the human genome and encoding nearly 900 genes .
C9orf21 demonstrates significant evolutionary conservation across mammalian species. Research using recombinant proteins and comparative analysis has shown:
The human and mouse orthologs share approximately 93% sequence identity
The human and rat orthologs share approximately 93% sequence identity
The bovine homolog (Q148E0) has been fully sequenced and characterized
This high degree of conservation suggests important functional roles that have been maintained throughout mammalian evolution. Researchers should consider this conservation when designing experiments using animal models, as findings may have translational relevance to human biology .
For detecting endogenous C9orf21/AAED1 in experimental settings, the following methodologies are recommended:
Western Blot:
Recommended antibody dilution: 1:300-5000
Sample preparation: Standard protein extraction with protease inhibitors
Expected molecular weight: Approximately 24.7 kDa
Immunohistochemistry (IHC):
IHC-Paraffin (IHC-P): Antibody dilution 1:50-200
IHC-Frozen (IHC-F): Antibody dilution 1:50-200
Antigen retrieval: Heat-mediated citrate buffer pH 6.0
Immunofluorescence (IF):
IF in cell culture (ICC): Antibody dilution 1:50-200
Counterstaining: DAPI for nuclear visualization
Fixation: 4% paraformaldehyde followed by permeabilization
For validation of antibody specificity, blocking experiments using recombinant protein fragments (such as Human AAED1 aa 84-142) at 100× molar excess compared to the antibody concentration is recommended .
When designing experiments to investigate C9orf21 function in vitro:
Gain-of-function approaches:
Transfection with expression plasmids containing tagged C9orf21 (options include Myc/DDK-tagged or GFP-tagged constructs)
Lentiviral transduction using vectors such as pLenti-C-mGFP-P2A-Puro for stable expression
Doxycycline-inducible expression systems for temporal control
Loss-of-function approaches:
siRNA or shRNA targeting multiple regions of C9orf21 mRNA
CRISPR/Cas9-mediated knockout using guide RNAs targeting early exons
Dominant-negative constructs based on critical functional domains
Functional readouts to consider:
Cell proliferation and viability assays (particularly relevant given association with cancer)
Metabolic analysis (glycolysis measurements, given evidence of metabolic modulation)
Oxidative stress response (given peroxiredoxin-like domain)
Protein-protein interaction studies to identify binding partners
When designing primers for qPCR validation, researchers should be aware of potential splice variants and ensure primers span exon-exon junctions .
For optimal storage and handling of recombinant C9orf21/AAED1 protein:
Storage conditions:
Store at -20°C to -80°C for long-term storage
For lyophilized form: 12 months stability at -20°C/-80°C
For liquid form: 6 months stability at -20°C/-80°C
Avoid repeated freeze-thaw cycles
For working aliquots: Store at 4°C for up to one week
Reconstitution protocol:
Centrifuge vial at 5,000×g for 5 minutes before opening
Reconstitute in deionized sterile water to 0.1-1.0 mg/mL
Add glycerol to a final concentration of 5-50% for long-term storage
Aliquot to minimize freeze-thaw cycles
Buffer considerations:
Typical storage buffers include:
100 mM glycine, 25 mM Tris-HCl, pH 7.3
TBS (pH 7.4) with 1% BSA, 0.02-0.03% Proclin300, and 50% Glycerol
For critical experiments, validation of protein activity after storage is recommended, especially if the protein is used for functional studies rather than as a control or standard .
Recent evidence suggests that AAED1 (C9orf21) modulates cellular metabolism, particularly glycolysis in cancer models. To investigate this role:
Experimental approaches:
Metabolic flux analysis: Use Seahorse XF Analyzer to measure extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) in cells with modulated C9orf21 expression
Glucose uptake assays: Measure uptake of labeled glucose (2-NBDG or tritiated 2-deoxyglucose) in control versus C9orf21-overexpressing or knockdown cells
Expression analysis of glycolytic enzymes: Perform qPCR and Western blot analyses of key glycolytic enzymes (HK2, PFKP, PKM2, LDHA) to determine if C9orf21 affects their expression
Metabolomics profiling: Conduct LC-MS-based metabolomics to comprehensively analyze changes in glycolytic intermediates and related metabolic pathways
A comprehensive experimental design should include multiple cell types and correlation with clinical samples to validate findings across biological contexts. The peroxiredoxin-like domain suggests potential involvement in redox regulation, which may link to metabolic functions through redox-sensitive metabolic enzymes .
It is important to note that C9orf21 and C9orf72 are distinct genes, though both are located on chromosome 9. This can lead to confusion in the literature:
Key distinctions:
C9orf72: Associated with hexanucleotide repeat expansions (GGGGCC) that cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD)
C9orf21 (AAED1/PRXL2C): Not directly implicated in repeat expansion disorders
Research considerations when studying neurodegeneration:
Differential expression analysis: Compare C9orf21 expression in post-mortem brain tissues from patients with C9orf72 expansions versus those without expansions
Potential functional interactions: Investigate whether C9orf21 interacts with C9orf72 or modifies pathological processes in C9orf72-related diseases
Co-expression networks: Use WGCNA (weighted gene co-expression network analysis) to determine if C9orf21 belongs to gene modules associated with neurodegeneration
Recent studies of cerebellar transcriptomes in C9orf72 expansion carriers have revealed widespread transcriptomic alterations. Investigating whether C9orf21 is part of these altered networks could provide insights into disease mechanisms .
Analysis of post-translational modifications (PTMs) of C9orf21/AAED1 requires systematic approaches:
Mass spectrometry-based strategies:
Sample preparation: Immunoprecipitate endogenous or tagged C9orf21 from cell lysates under conditions that preserve PTMs (phosphatase/deacetylase inhibitors)
MS analysis options:
Shotgun proteomics for broad PTM identification
Targeted MS for specific modification sites
TMT-based quantitative proteomics for comparing PTM levels across conditions
Isotopic labeling: Use of C13 and N15-labeled recombinant protein as an internal standard for accurate quantification
Site-specific mutation analysis:
Identify putative modification sites through in silico prediction tools
Generate site-specific mutants (e.g., S→A for phosphorylation, K→R for ubiquitination/acetylation)
Compare functional outcomes between wild-type and mutant proteins
PTMs of particular interest:
Phosphorylation (multiple potential Ser/Thr/Tyr sites)
Redox modifications (given peroxiredoxin domain)
Ubiquitination (potential regulation of protein stability)
Researchers should consider using C13 and N15-labeled recombinant protein as a standard for mass spectrometry experiments, which allows precise quantification of modifications across experimental conditions .
Research has demonstrated that C9orf21/AAED1 expression is altered in certain cancers, with particular evidence in gastric cancer:
Expression patterns:
Increased expression observed in gastric cancer tissues compared to adjacent normal tissues
Expression levels correlate with clinical parameters including tumor size and stage
Functional implications:
Proliferation: AAED1 modulates cancer cell proliferation, with knockdown resulting in reduced proliferation rates
Metabolism: AAED1 affects glycolytic metabolism in cancer cells, potentially contributing to the Warburg effect
Molecular mechanisms: May involve interaction with key metabolic enzymes or signaling pathways that regulate cell growth
Experimental validation approaches:
Colony formation assays
Cell cycle analysis by flow cytometry
In vivo tumor xenograft models
Correlation analysis with clinical outcomes in patient cohorts
Researchers investigating C9orf21/AAED1 in cancer should consider both its direct effects on cellular processes and its potential as a biomarker for disease progression or therapeutic response .
When investigating C9orf21/AAED1 in relation to the blood-brain barrier (BBB), several specialized methodological approaches should be considered:
In vitro BBB models:
Transwell co-culture systems: Human brain microvascular endothelial cells (hBMECs) with astrocytes and/or pericytes
Microfluidic organ-on-chip platforms: For dynamic flow conditions that better mimic physiological BBB
Measurement parameters: TEER (transendothelial electrical resistance), permeability coefficients for model compounds
Ex vivo approaches:
Isolated brain microvessels: For protein/mRNA extraction and analysis of C9orf21 expression
Brain microvessel endothelial cell isolation: For proteomic analysis, as demonstrated in studies with C9orf72 models
In vivo approaches:
In situ transcardiac brain perfusion: Has been successfully used to study BBB transport in C9orf72 models
Tandem Mass Tag (TMT)-proteomics: For analysis of isolated brain microvascular endothelial cells (BMECs)
Data analysis considerations:
Comparison between disease models (e.g., C9orf72 expansion models) and wild-type controls
Correlation with BBB transport markers (e.g., glucose transporter expression)
Functional enrichment analysis of proteomic data
These methodologies have been successfully applied in C9orf72 studies and could be adapted to investigate C9orf21/AAED1 in BBB function and dysfunction .
| BBB Property | Measurement Technique | Relevance to C9orf21 Research |
|---|---|---|
| Passive diffusion | 14C-sucrose, 3H-diazepam permeability | Baseline BBB integrity assessment |
| Transporter function | 3H-2-deoxy-D-glucose transport | Potential metabolic regulation |
| Tight junction integrity | Zonula occludens-1 quantification | BBB structural analysis |
| Efflux transport | 3H-digoxin (P-gp substrate) | Drug delivery considerations |
When encountering contradictory findings regarding C9orf21/AAED1 across different experimental systems, researchers should consider:
Sources of experimental variation:
Cell/tissue type differences: C9orf21 may have context-dependent functions
Expression level differences: Overexpression systems may produce artifacts not present at endogenous levels
Tag interference: Different protein tags (Myc/DDK, GFP, etc.) may affect protein function
Species differences: Despite high conservation (93% between human and mouse/rat), species-specific functions may exist
Resolution strategies:
Cross-validation: Employ multiple experimental approaches (e.g., both gain- and loss-of-function)
Physiological relevance: Prioritize findings from systems with endogenous expression levels
Rescue experiments: Perform complementation studies to validate specificity
Dose-response relationships: Determine if effects are dependent on expression levels
Temporal considerations: Assess acute versus chronic effects of modulating C9orf21
Integration framework:
Develop a working model that accommodates seemingly contradictory findings
Consider potential context-specific regulatory mechanisms
Validate key findings in primary cells or tissues when possible
This systematic approach helps reconcile disparate findings and builds a more comprehensive understanding of C9orf21/AAED1 biology .
While C9orf21 is distinct from C9orf72 (which is known for hexanucleotide repeat expansions), the methodological considerations for repeat-primed PCR are valuable for researchers studying genomic regions:
Primer design considerations:
Specificity: Design primers that uniquely target C9orf21 with minimal off-target binding
GC content: Optimize GC content (40-60%) to ensure efficient amplification
Annealing temperature: Empirically determine optimal temperature for specificity
Amplicon size: Keep amplicons under 200bp for optimal efficiency
PCR optimization parameters:
Mastermix components: Consider specialized mixes for difficult templates (high GC content)
Additives: 5M Betaine can improve amplification of difficult templates
Cycling conditions:
Initial denaturation: 95°C for 10 minutes
Two-stage cycling with gradually increasing extension times
Final extension at 72°C for 7 minutes
Controls and validation:
Include positive and negative controls in each experiment
Verify PCR products by sequencing to confirm target specificity
Consider alternative methods (qPCR, digital PCR) for validation
The protocols used for C9orf72 repeat expansion detection can provide methodological guidance, though they should be adapted specifically for C9orf21 genomic regions of interest .
Recombinant C9orf21/AAED1 protein serves as an important control in various experimental settings:
Antibody validation:
Blocking experiments: Pre-incubate antibody with recombinant protein (100× molar excess) for 30 minutes at room temperature before application to samples
Western blot positive control: Load 10-50 ng of recombinant protein alongside experimental samples
ELISA standard curve: Use serial dilutions (0.1-1000 ng/mL) for quantitative assays
Mass spectrometry applications:
C13 and N15-labeled protein: Use as internal standard for absolute quantification
Peptide identification: Generate theoretical peptide maps from recombinant sequence
Post-translational modification analysis: Compare modification patterns between recombinant and endogenous protein
Functional assays:
Enzyme activity baseline: Establish baseline activity for comparison with mutant variants
Protein-protein interaction controls: Use as bait protein in pull-down experiments
Structural studies: Purified protein for crystallography or NMR studies
When using recombinant protein, researchers should verify protein quality through SDS-PAGE (>85% purity) and consider potential differences from endogenous protein (such as tag effects or lack of post-translational modifications) .
| Application | Recommended Amount | Key Considerations |
|---|---|---|
| Western blot blocking | 100× molar excess | Pre-incubate 30 min at RT |
| Positive control | 10-50 ng/lane | Include on each blot |
| ELISA standard | 0.1-1000 ng/mL | Prepare fresh dilutions |
| MS standard | 50 μg/mL | C13/N15-labeled for quantification |
When implementing co-expression network analyses to identify functional associations of C9orf21/AAED1:
Data preparation and quality control:
Expression normalization: Use appropriate normalization methods (e.g., TMM, quantile normalization)
Batch effect correction: Apply ComBat or similar methods if data comes from multiple sources
Outlier detection: Remove samples with extreme expression patterns
Covariate adjustment: Generate residual expression values adjusted for relevant covariates (age, sex, RIN)
Network construction parameters:
Correlation method: Use biweight mid-correlation ("bicor") for robustness against outliers
Signed hybrid network: Preserves direction of correlations
Power threshold: Set to maintain scale-free topology (typically 6-12)
Module detection: Apply dynamic tree cutting with merge height of 0.4 and minimum module size of 30
Functional interpretation:
Module eigengene analysis: Correlate with phenotypic traits or experimental conditions
Module membership calculation: Determine strength of association between C9orf21 and each module
Hub gene identification: Identify central genes in modules containing C9orf21
Pathway enrichment analysis: Determine biological processes represented in C9orf21-containing modules
Visualization and validation:
Network visualization: Use Cytoscape (v3.8.2 or newer) to create interactive network graphs
Cross-validation: Split data into discovery and validation cohorts
Experimental validation: Test predicted interactions through co-immunoprecipitation or functional assays