AFG3L2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on your location and the shipping method you choose. Please consult your local distributor for specific delivery times.
Synonyms
AFG3 (ATPase family gene 3; yeast) like 2 antibody; AFG3 ATPase family gene 3 like 2 (yeast) antibody; AFG3 ATPase family gene 3 like 2 antibody; AFG3 like protein 2 antibody; AFG3-like protein 2 antibody; AFG32_HUMAN antibody; AFG3L2 antibody; ATPase family gene 3 like 2 antibody; ATPase family gene 3 yeast antibody; EC 3.4.24.- antibody; FLJ25993 antibody; Paraplegin like protein antibody; Paraplegin-like protein antibody; SCA28 antibody; Spinocerebellar ataxia 28 antibody
Target Names
AFG3L2
Uniprot No.

Target Background

Function
AFG3L2 is an ATP-dependent protease essential for axonal and neuronal development. Within neurons, it mediates the degradation of SMDT1/EMRE before its assembly with the uniporter complex, limiting the availability of SMDT1/EMRE for MCU assembly and promoting efficient assembly of gatekeeper subunits with MCU. AFG3L2 is also required for paraplegin (SPG7) maturation. After cleavage by mitochondrial-processing peptidase (MPP), it converts paraplegin into a proteolytically active mature form. It is further involved in the maturation of PINK1 to its 52 kDa mature form following cleavage by MPP. Finally, AFG3L2 plays a role in regulating the OMA1-dependent processing of OPA1.
Gene References Into Functions
  1. Deletion of AFG3L2 has been associated with spinocerebellar ataxia type 28 (SCA28) in the context of multiple genomic anomalies. PMID: 25251419
  2. AOA2 with myoclonus has been linked to mutations in SETX and AFG3L2. PMID: 25927548
  3. Research suggests that AFG3L2 mutations are another significant cause, albeit rare, of a late-onset ataxic PEO phenotype resulting from a disturbance of mtDNA maintenance. PMID: 25420100
  4. StAR proteolysis is carried out by at least two mitochondrial proteases: the matrix LON protease and the inner membrane complexes of the metalloproteases AFG3L2 and AFG3L2:SPG7/paraplegin. PMID: 24422629
  5. A novel AFG3L2 mutation has been reported in a patient with slowly progressive ataxia and a positive family history. PMID: 24293060
  6. Identification of a partial AFG3L2 deletion and subsequent functional studies indicate loss of function as the most likely disease mechanism. PMID: 24814845
  7. Both full-length and truncated COX1 proteins physically interact with AFG3L2. PMID: 22252130
  8. These findings broaden the phenotype associated with AFG3L2 mutations and suggest that AFG3L2-related disease should be considered in the differential diagnosis of spastic ataxias. PMID: 22022284
  9. Mutations in SCA28 are associated with amino acid changes in evolutionarily conserved residues of the alleged SCA28 gene, indicating SCA28 as the sixth recognized SCA genotype caused by point mutations. PMID: 21827917
  10. Studies further confirm both the involvement of AFG3L2 gene in Spinocerebellar ataxia type 28 (SCA28) and the presence of a mutational hotspot in exons 15-16. PMID: 20725928
  11. A study on spinocerebellar ataxia type 28 patients identified a novel missense mutation at an evolutionarily conserved amino-acid position; amino-acid exchange p.E700K was detected in a 4-generation family and was not observed in control chromosomes. PMID: 20354562
  12. Research identifies AFG3L2 as a novel cause of dominant neurodegenerative disease and indicates a previously unknown role for this component of the mitochondrial protein quality control machinery in protecting the human cerebellum against neurodegeneration. PMID: 20208537
  13. An intersubunit signaling network coordinates ATP hydrolysis by the m-AAA protease AGG3L2. PMID: 19748354

Show More

Hide All

Database Links

HGNC: 315

OMIM: 604581

KEGG: hsa:10939

STRING: 9606.ENSP00000269143

UniGene: Hs.726355

Involvement In Disease
Spinocerebellar ataxia 28 (SCA28); Spastic ataxia 5, autosomal recessive (SPAX5)
Protein Families
AAA ATPase family; Peptidase M41 family
Subcellular Location
Mitochondrion. Mitochondrion inner membrane; Multi-pass membrane protein.
Tissue Specificity
Ubiquitous. Highly expressed in the cerebellar Purkinje cells.

Q&A

What is AFG3L2 and why is it important in mitochondrial research?

AFG3L2 (AFG3 ATPase family gene 3-like 2) is the catalytic subunit of the m-AAA protease, an ATP-dependent proteolytic complex in the mitochondrial inner membrane. This protein possesses dual functionality with both ATPase and protease activities. The ATPase activity unfolds substrate proteins, threading them into the internal proteolytic cavity for hydrolysis into small peptide fragments .

AFG3L2 is critical for several essential cellular processes:

  • Proteostasis of inner mitochondrial membrane proteins

  • Quality control by degrading mistranslated or misfolded polypeptides

  • Processing and maturation of mitochondrial proteins (including MRPL32/bL32m, PINK1, and SP7)

  • Axonal and neuronal development, particularly in Purkinje cells

AFG3L2 forms either homo-oligomeric complexes or hetero-oligomeric complexes with SPG7 (paraplegin), assembling into hexameric structures within the inner mitochondrial membrane . Defects in AFG3L2 are causally linked to spinocerebellar ataxia type 28 (SCA28) and spastic ataxia autosomal recessive type 5 (SPAX5) .

What are the typical molecular weights observed for AFG3L2 in Western blot analysis?

While the calculated molecular weight of AFG3L2 is approximately 89 kDa , researchers should note several important considerations when analyzing Western blot results:

  • The observed molecular weight is typically around 80 kDa in most tissues and cell lines

  • A truncated 65 kDa form of AFG3L2 has been reported in some experimental systems

  • Post-translational modifications may affect migration patterns

  • In disease models carrying AFG3L2 mutations, proteomic analysis has shown approximately 60% residual expression compared to wild-type cells

Researchers should validate bands using appropriate positive controls such as HeLa cells, Jurkat cells, or mouse brain tissue, which consistently show strong AFG3L2 expression .

What applications are AFG3L2 antibodies validated for?

AFG3L2 antibodies have been validated for multiple applications across different experimental paradigms:

ApplicationValidated Antibody TypesNotes
Western Blot (WB)Rabbit polyclonal, Mouse monoclonalDilution ranges: 1:100-1:16000 depending on antibody
Immunoprecipitation (IP)Rabbit polyclonalTypically 0.5-4.0 μg antibody for 1.0-3.0 mg total protein
Immunohistochemistry (IHC)Rabbit polyclonalDilution ranges: 1:50-1:400, may require antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0
Immunofluorescence (IF)/ICCRabbit polyclonal, Mouse monoclonalDilution ranges: 1:10-1:100
Flow Cytometry (FC)Mouse monoclonalPrimarily for intracellular detection
ELISARabbit polyclonal, Mouse monoclonalVarious formats supported

Validation has been performed across human, mouse, and rat samples, with some antibodies also showing reactivity with pig and rabbit samples .

How should researchers design experiments to study AFG3L2 mutations associated with neurological disorders?

When investigating AFG3L2 mutations associated with disorders like SCA28 and SPAX5, consider this multi-faceted experimental approach:

  • Cell Model Selection:

    • Immortalized lymphoblastoid cell lines provide a robust in vitro disease model for studying biochemical fingerprints relevant to neuronal pathophysiology

    • Neuronal cell lines (particularly those derived from Purkinje cells) are ideal given AFG3L2's high expression in cerebellar Purkinje cells

  • Multi-omics Integration:

    • Combine proteomics, lipidomics, and targeted metabolomics analyses to comprehensively characterize molecular alterations induced by AFG3L2 variants

    • Proteomic analysis can reveal residual expression levels of mutant AFG3L2 proteins and identify dysregulated interaction partners such as MCU

  • Mitochondrial Function Assessment:

    • Examine mitochondrial fragmentation and displacement within dendritic structures

    • Assess protein synthesis and ribosomal assembly within mitochondria

    • Measure oxidative stress and inflammatory markers associated with AFG3L2 deficiency

  • Phenotypic Characterization:

    • Monitor axonal development and myelination

    • Evaluate interactions between neurons and glial cells, particularly astrocytes which are also affected by AFG3L2 deficiency

This integrated approach allows researchers to delineate the complex molecular mechanisms linking mitochondrial dysfunction to cellular architecture and survival mechanisms in AFG3L2-related disorders.

What are the critical considerations for optimizing immunoprecipitation protocols to study AFG3L2 protein-protein interactions?

Successful immunoprecipitation of AFG3L2 and its protein interaction partners requires careful optimization:

  • Antibody Selection:

    • Choose antibodies specifically validated for IP applications (e.g., ab241158 has been validated at 6 μg per reaction)

    • Consider using antibodies targeting different epitopes to confirm interactions

  • Lysis Buffer Composition:

    • NETN lysis buffer has been successfully used for AFG3L2 immunoprecipitation

    • Gentle detergents are critical to preserve native protein-protein interactions in mitochondrial membrane complexes

  • Protocol Optimization:

    • For co-immunoprecipitation studies, typically use 0.5-4.0 μg antibody for 1.0-3.0 mg of total protein lysate

    • Include appropriate controls such as non-specific IgG IP to identify non-specific binding

  • Detection Strategy:

    • Western blot analysis using the same or different AFG3L2 antibody (typically at 0.1-0.4 μg/ml concentration)

    • Consider chemiluminescence with exposure times around 30 seconds for optimal signal detection

  • Validation of Interactions:

    • Confirm known interactions (e.g., with SPG7/paraplegin) before investigating novel binding partners

    • Consider proximity ligation assays as complementary techniques to validate interactions identified by IP

How can researchers effectively use proteomics to characterize AFG3L2 expression and its interactome?

Advanced proteomic approaches offer powerful insights into AFG3L2 function and dysregulation:

  • Coverage Optimization:

    • Comprehensive proteomic analysis can achieve excellent coverage of the AFG3L2 protein sequence (e.g., 21 unique tryptic peptides have been used to cover significant portions of the protein)

  • Quantification Methodology:

    • Data-independent-acquisition (DIA) mode provides reliable protein quantification

    • For optimal quantification, use the 3 highest abundant peptides for each protein

  • Dynamic Range Considerations:

    • Proteomic analyses can cover a dynamic range of nearly nine orders of magnitude, allowing detection of both abundant and rare proteins in the same experiment

  • Statistical Analysis:

    • Apply appropriate statistical methods to identify differentially abundant proteins (DAPs)

    • Consider both fold change (typically log2 FC >1 or <-1) and statistical significance (p<0.05) when identifying dysregulated proteins

  • Functional Categorization:

    • Use tools like Proteomaps to categorize differentially abundant proteins according to their functional roles

    • Separate analysis of upregulated versus downregulated proteins can reveal distinct cellular processes affected by AFG3L2 dysfunction

This approach allows researchers to not only quantify AFG3L2 expression levels but also understand the broader impact of AFG3L2 mutations or deficiency on the cellular proteome.

What are the recommended protocols for detecting AFG3L2 in Western blot experiments?

For optimal Western blot detection of AFG3L2, researchers should follow these methodological guidelines:

  • Sample Preparation:

    • NETN lysis buffer has been successfully used for sample preparation

    • Typical loading amounts are approximately 50 μg of total cell lysate per lane

  • Antibody Selection and Dilution:

    • Rabbit polyclonal antibodies: typically used at 0.1-0.4 μg/ml or dilutions of 1:2000-1:16000

    • Mouse monoclonal antibodies: follow manufacturer's recommended dilutions

  • Detection Systems:

    • ECL (Enhanced Chemiluminescence) technique provides good results

    • Typical exposure times of approximately 30 seconds are sufficient for visualization

  • Expected Results:

    • Primary band at approximately 80 kDa in most tissue/cell types

    • Positive controls: HeLa cells, HEK-293T cells, Jurkat cells, mouse brain tissue, and mouse skeletal muscle have all been validated as positive controls for AFG3L2 detection

  • Troubleshooting:

    • If the signal is weak, consider longer primary antibody incubation times or increased antibody concentration

    • For high background, increase washing steps or adjust blocking conditions

This optimized protocol consistently yields specific detection of AFG3L2 across various human and rodent cell lines and tissues.

What are the best practices for immunohistochemical detection of AFG3L2 in tissue samples?

For successful immunohistochemical staining of AFG3L2 in tissue sections:

  • Tissue Preparation:

    • Formalin-fixed, paraffin-embedded (FFPE) sections are commonly used

    • Section thickness of 5-7 μm is typically appropriate

  • Antigen Retrieval:

    • TE buffer at pH 9.0 is the preferred method for antigen retrieval

    • Alternatively, citrate buffer at pH 6.0 can be used if TE buffer yields suboptimal results

    • Heat-induced epitope retrieval (pressure cooker or microwave) is generally more effective than enzymatic methods

  • Antibody Dilution and Incubation:

    • Recommended dilution ranges from 1:50 to 1:400 depending on the specific antibody

    • Overnight incubation at 4°C often yields better results than shorter incubations at room temperature

  • Detection System:

    • HRP-conjugated secondary antibodies with DAB substrate provide good contrast

    • For fluorescent detection, consider Alexa Fluor-conjugated secondary antibodies

  • Validation Controls:

    • Human kidney tissue has been successfully used as a positive control

    • Include no-primary-antibody controls to assess background staining

    • When available, use tissue from AFG3L2 knockout models as negative controls

  • Expected Staining Pattern:

    • Predominantly mitochondrial, with strong staining in tissues with high mitochondrial content

    • Particularly strong expression in cerebellar Purkinje cells

These optimized protocols allow for specific visualization of AFG3L2 expression patterns across different tissue types and disease models.

How should researchers interpret variations in AFG3L2 expression levels across different experimental systems?

When analyzing AFG3L2 expression data, researchers should consider these interpretative guidelines:

  • Tissue-Specific Expression Patterns:

    • AFG3L2 is highly and selectively expressed in human cerebellar Purkinje cells

    • Strong expression is also observed in tissues with high metabolic demands

  • Subcellular Localization Considerations:

    • AFG3L2 should localize to mitochondria, specifically the inner mitochondrial membrane

    • Altered localization may indicate mitochondrial dysfunction or experimental artifacts

  • Expression in Disease Models:

    • SPAX5 patient-derived cells show approximately 60% residual expression of AFG3L2 compared to wild-type

    • Missense variants may be expressed but functionally compromised

  • Interpreting Multiple Isoforms:

    • The presence of both 80 kDa and 65 kDa forms requires careful interpretation

    • The truncated 65 kDa form may represent alternative processing or degradation products

  • Quantification Approaches:

    • For accurate comparative quantification, normalize AFG3L2 levels to appropriate housekeeping proteins

    • Consider multiple normalization methods when comparing across diverse tissue types

Understanding these patterns allows researchers to effectively distinguish between normal biological variation and pathological changes in AFG3L2 expression.

What are the key findings from multi-omics studies of AFG3L2 mutations and their implications for future research?

Recent multi-omics approaches have revealed important insights about AFG3L2 dysfunction:

  • Proteomic Alterations:

    • AFG3L2 mutations result in significant proteome-wide dysregulation

    • Studies have identified 63 differentially abundant proteins (47 increased, 16 decreased) in AFG3L2 mutant cells

    • Key binding partners, such as MCU (mitochondrial calcium uniporter), show specific dysregulation

  • Functional Pathway Analysis:

    • Proteomaps analysis reveals distinct functional categories for upregulated versus downregulated proteins

    • Changes in mitochondrial protein composition reflect compensatory mechanisms and direct effects of AFG3L2 dysfunction

  • Metabolic Consequences:

    • Integration of proteomics with lipidomics and targeted metabolomics analyses shows broader metabolic disturbances

    • These changes provide potential biomarkers for monitoring disease progression or therapeutic responses

  • Research Implications:

    • Immortalized lymphoblastoid cell lines serve as valuable disease models for studying biochemical alterations relevant to neuronal pathophysiology

    • Multi-omics approaches are essential for comprehensively understanding the molecular consequences of AFG3L2 mutations

    • Future research should focus on validating identified dysregulated pathways in neuronal models and exploring their potential as therapeutic targets

These findings emphasize the importance of integrated analytical approaches in understanding the complex molecular mechanisms underlying AFG3L2-related neurological disorders.

What strategies can resolve common challenges in detecting AFG3L2 in challenging sample types?

When working with difficult tissue samples or experimental conditions:

  • Low Signal Issues:

    • For neuronal tissues with complex extracellular matrix, increase antigen retrieval time and optimize buffer pH

    • Consider signal amplification systems such as tyramide signal amplification for IHC/IF applications

    • For Western blots, membrane transfer conditions may need optimization for this large protein (80-89 kDa)

  • High Background Problems:

    • Implement more stringent blocking with 5% BSA or specialized blocking reagents

    • For immunofluorescence, include an autofluorescence quenching step

    • Consider monoclonal antibodies for higher specificity in challenging sample types

  • Multiple Band Detection:

    • Validate bands using positive and negative controls to distinguish specific signals

    • Consider pre-absorbing antibodies with recombinant protein to confirm specificity

    • Use gradient gels for better separation of protein isoforms

  • Sample Preparation Optimization:

    • For mitochondrial membrane proteins like AFG3L2, detergent selection is critical (mild non-ionic detergents generally preserve structure)

    • Fresh samples yield better results than frozen samples for many applications

    • Protease inhibitor cocktails are essential to prevent degradation during preparation

These advanced troubleshooting strategies can significantly improve detection and analysis of AFG3L2 across diverse experimental contexts.

How can researchers verify antibody specificity for AFG3L2 in knockout/knockdown studies?

Rigorous validation of AFG3L2 antibody specificity requires:

  • Genetic Manipulation Controls:

    • CRISPR/Cas9 knockout cells provide the gold standard for antibody validation

    • siRNA or shRNA knockdown samples can serve as partial validation controls

    • Overexpression systems with tagged AFG3L2 offer complementary validation approaches

  • Western Blot Validation:

    • Compare samples with different AFG3L2 expression levels side-by-side

    • Demonstrate proportional signal reduction corresponding to knockdown efficiency

    • Verify absence of the specific band in knockout samples while housekeeping proteins remain detectable

  • Immunofluorescence Confirmation:

    • Demonstrate loss of mitochondrial staining pattern in knockout/knockdown cells

    • Co-localization with mitochondrial markers should be reduced or eliminated

    • Quantify signal intensity across multiple cells and experiments for statistical validation

  • Complementation Studies:

    • Re-expression of AFG3L2 in knockout cells should restore the signal

    • Rescue experiments with wild-type but not mutant forms provide additional specificity confirmation

  • Cross-Validation:

    • Compare results from multiple antibodies targeting different epitopes of AFG3L2

    • Correlation between protein detection methods (WB, IF, IP) strengthens validation

This comprehensive validation approach ensures reliable and specific detection of AFG3L2 in experimental systems.

How can AFG3L2 antibodies facilitate research into mitochondrial dynamics and quality control mechanisms?

AFG3L2 antibodies enable several cutting-edge research applications:

  • Super-Resolution Microscopy:

    • High-quality antibodies compatible with techniques like STORM or STED can reveal detailed mitochondrial substructure

    • Co-localization studies with other mitochondrial quality control components provide insights into functional organization

  • Live-Cell Imaging:

    • Membrane-permeable fluorescently-labeled antibody fragments can potentially monitor AFG3L2 dynamics in living cells

    • Correlate AFG3L2 localization with mitochondrial fragmentation events in real-time

  • Proximity Labeling Approaches:

    • AFG3L2 antibodies can be used to validate results from BioID or APEX2 proximity labeling experiments

    • These techniques can identify novel interaction partners within the mitochondrial quality control network

  • Single-Cell Analysis:

    • Antibody-based detection in flow cytometry or mass cytometry (CyTOF) enables quantification of AFG3L2 levels at single-cell resolution

    • Correlate AFG3L2 expression with other mitochondrial markers to identify heterogeneous cellular responses

  • Therapeutic Development:

    • Screening for compounds that modulate AFG3L2 expression or activity

    • Monitoring AFG3L2 levels as biomarkers for mitochondrial dysfunction in neurodegenerative disorders

These emerging applications position AFG3L2 antibodies as essential tools for advancing our understanding of fundamental mitochondrial biology and pathological mechanisms.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.