SACS Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery information.
Synonyms
DnaJ homolog subfamily C member 29 antibody; DNAJC29 antibody; SACS antibody; SACS_HUMAN antibody; Sacsin antibody; Spastic ataxia of Charlevoix-Saguenay antibody
Target Names
SACS
Uniprot No.

Target Background

Function
Sacsin is a co-chaperone that acts as a regulator of the Hsp70 chaperone machinery. It is believed to play a role in the processing of other ataxia-linked proteins.
Gene References Into Functions
  1. Our findings confirm the broad clinical spectrum associated with SACS mutations, including pure polyneuropathy without characteristic clinical and brain imaging manifestations of autosomal recessive spastic ataxia of Charlevoix-Saguenay. PMID: 30460542
  2. This study presents the first Polish family with a comprehensive clinical and neuropsychological assessment, harboring two novel mutations in the SACS gene. PMID: 28843771
  3. The twins described by Fitzsimmons had heterozygous mutations in the SACS gene, responsible for autosomal recessive spastic ataxia of Charlevoix Saguenay, along with a heterozygous mutation in the TRPS1 gene, responsible for Trichorhinophalangeal syndrome type 1A. A TBL1XR1 mutation was identified in the patient described in 2009 as contributing to his cognitive impairment and autistic features. PMID: 27133561
  4. This study provides a potential genetic diagnosis for the patient and expands the spectrum of SACS mutations. PMID: 28658676
  5. A genetic diagnosis was established in six families with autosomal recessive HSP (SPG11 in three families and TFG/SPG57, SACS and ALS2 in one family each). A heterozygous mutation in a gene involved in an autosomal dominant HSP (ATL1/SPG3A) was also identified in one additional family. PMID: 27601211
  6. The results support the notion that the HEPN domain contributes to the functional activity of sacsin by binding to nucleotides or other multiply charged anionic compounds in neurons. PMID: 26366743
  7. Various SACS mutations have functional consequences on the mitochondrial compartment in ARSACS patients. PMID: 26288984
  8. This study reports an Italian family affected by an autosomal recessive form of hereditary spastic paraplegia (HSP) and peripheral neuropathy caused by a novel mutation in the SACS gene. PMID: 23800155
  9. To clarify the segregation pattern of the mutations found in this family, after excluding somatic mosaicism for the specific mutations, the SACS gene was fully reanalyzed. PMID: 24164681
  10. Whole-exome sequencing identified a hemizygous novel spastic ataxia of Charlevoix-Saguenay (SACS) stop-codon mutation in 2 brothers. PMID: 24180463
  11. Abnormal retinal thickening is a common feature in patients with SACS mutation phenotype. PMID: 24457356
  12. Widespread tissue damage may be associated with extensive loss of sacsin protein in the brain and could explain a wide range of progressive neurologic abnormalities in patients with spastic ataxia of Charlevoix-Saguenay. PMID: 23598833
  13. A novel missense mutation in sacsin, p.Arg272His, was identified in a patient with sacsin-related spastic ataxia. PMID: 23338241
  14. The relative position of mutations in subrepeats will variably influence sacsin dysfunction. PMID: 23280630
  15. A new mutation in the SACS gene was identified in a patient with Autosomal recessive cerebellar ataxia. PMID: 23043354
  16. This study demonstrated that 16 novel SACS gene mutations in recessive spastic ataxia of Charlevoix-Saguenay showed supratentorial and pontine abnormalities. PMID: 22816526
  17. This study compares the GHKL-type-ATPase domain of this human protein to that of related plant, protozoan, yeast and bacterial proteins. PMID: 19880797
  18. Novel compound heterozygous frameshift mutations were detected in the SACS gene in two siblings with a sensorimotor neuropathy, ataxia, and spasticity. PMID: 22751902
  19. As more SACS mutations are identified worldwide, the clinical spectrum of 'sacsinopathies' will expand. PMID: 17853117
  20. This study describes two unrelated Autosomal recessive spastic ataxia of Charlevoix Saguenay patients from central Italy carrying two novel mutations in SACS. PMID: 22805644
  21. This study demonstrated that Autosomal recessive spastic ataxia of Charlevoix-Saguenay is associated with compound heterozygotes for nonsense mutations of the SACS gene. PMID: 21745802
  22. This study provides novel insights into the oligomerization state of sacsin and functions that are lost in mutations that cause ARSACS. PMID: 21507954
  23. Data shows that uniparental isodisomy of the paternal chromosome 13 carrying the mutated SACS gene played an etiologic role in a case of the disease. PMID: 20852969
  24. This study expands the ARSACS phenotype and the underlying genetic spectrum of SACS mutations. Patients with ARSACS are more common than previously known and may be underdiagnosed due to late onset age and unusual presentation. PMID: 20876471
  25. These data indicate that sacsin repeating regions necessitate nucleotide hydrolysis for their function, provided by the common Hsp90 ATPase domain, which may give rise to a novel activity related to protein quality control. PMID: 20488193
  26. Our results expand the genotype phenotype correlation of mutations in the sacsin gene in ataxia patients. PMID: 19892370
  27. This study reports a reliable and inexpensive method to detect more than 95% of the ARSACS disease alleles. PMID: 11788093
  28. The authors report a new mutation (1859insC), leading to a frameshift with a premature termination of the gene product sacsin, in two sisters from consanguineous parents. The phenotype is similar to previously described patients with ARSACS. PMID: 14718706
  29. The authors identified three new SACS mutations in two Italian patients whose phenotype closely matches that of Quebec cases, but without retinal striation. PMID: 14718707
  30. A homozygous missense mutation (T7492C) in the SACS gene, which resulted in the substitution of arginine for tryptophan at amino acid residue 2498 (W2498R) was identified in two sibling Japanese early onset spastic ataxia patients. PMID: 14718708
  31. This study analyzes SACS mutations in autosomal recessive spastic ataxia of Charlevoix-Saguenay families. PMID: 15156359
  32. The authors describe two Japanese siblings with autosomal recessive spastic ataxia of Charlevoix-Saguenay without spasticity, typically a core feature of this disorder. They had a novel homozygous missense mutation (T987C) of the SACS gene. PMID: 15985586
  33. The authors report here identical twin sisters with novel compound heterozygous mutations (c.[2951_2952delAG]+[3922delT]) in the SACS gene. PMID: 16198375
  34. This study describes a Japanese autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) patient with a compound heterozygous mutation in a new exon of the SACS gene. PMID: 16606928
  35. This study reports Japanese siblings with a new missense mutation (C922T, L308F) in exon 7 of SACS. PMID: 17290461
  36. This study describes four patients in a Belgian family with autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS). A novel homozygous missense mutation of the SACS gene was identified in this family. PMID: 17716690
  37. This study describes both point mutation and deletion associated with autosomal recessive spastic ataxia of Charlevoix-Saguenay. PMID: 18398442
  38. In a Dutch cohort of 43 index patients with ataxia onset before age 25, 16 index patients (total 23 patients) with mutations in the SACS gene were identified. Nine of them had homozygous mutations, and seven of them had compound heterozygous mutations. PMID: 18465152
  39. This study reports a clinical and genetic analysis of a Japanese family with ARSACS with novel compound heterozygous mutations in the SACS gene (N161fsX175, L802P). The phenotype is similar to that of previously reported ARSACS patients. PMID: 18484239
  40. Results report the identification of an unconventional SACS mutation, a large-scale deletion sized approximately 1.5 Mb encompassing the whole gene, in two unrelated patients. PMID: 19031088

Show More

Hide All

Database Links

HGNC: 10519

OMIM: 270550

KEGG: hsa:26278

STRING: 9606.ENSP00000371729

UniGene: Hs.159492

Involvement In Disease
Spastic ataxia Charlevoix-Saguenay type (SACS)
Subcellular Location
Cytoplasm. Note=Predominantly cytoplasmic, a small portion is present in the nucleus and also shows a partial mitochondrial overlap with the mitochondrial marker Hsp60.
Tissue Specificity
Highly expressed in the central nervous system. Also found in skeletal muscle and at low levels in pancreas.

Q&A

What is the SACS gene and sacsin protein?

The SACS gene encodes sacsin, a large protein associated with Autosomal Recessive Spastic Ataxia of Charlevoix-Saguenay (ARSACS), a neurodegenerative disorder. Research using sacsin antibodies has revealed that SACS mutations can dramatically reduce or completely eliminate full-length sacsin protein expression in patient cells, regardless of whether the mutations are missense or frameshift variations . Understanding sacsin's normal function and pathological alterations is critical for ARSACS research, with antibodies serving as essential tools for protein detection and characterization.

What types of SACS antibodies are available for research?

Several types of SACS antibodies are available for research applications, each with distinct characteristics:

  • C-terminal anti-sacsin antibodies (AbC): Commercially available antibodies recognizing the C-terminal portion of sacsin protein .

  • N-terminal anti-sacsin antibodies (AbN): Custom-developed antibodies targeting the N-terminal region, such as those raised against amino acids 1-728 of sacsin .

  • Commercial antibodies: Products like Human SACS Antibody (AF8014) from R&D Systems targeting specific sacsin regions .

Like other research antibodies, SACS antibodies can be polyclonal (derived from multiple B-cell lineages and recognizing various epitopes) or monoclonal (from a single B-cell lineage recognizing a single epitope) . The selection between these types depends on the specific research question and experimental approach.

How do SACS antibodies function in experimental systems?

SACS antibodies function as molecular recognition tools that bind specifically to sacsin protein or its fragments. These immunoglobulins are typically produced by immunizing host animals with sacsin peptides or recombinant protein fragments . The resulting antibodies can recognize their target in various experimental contexts, including denatured conditions (Western blotting), native conditions (immunoprecipitation), or in fixed tissues (immunohistochemistry). The specificity of these antibodies can be validated using controls such as CRISPR-Cas9-engineered SACS knockout cell lines, which should show no signal when probed with a specific SACS antibody .

How should researchers select the appropriate SACS antibody for specific applications?

Selecting the appropriate SACS antibody requires consideration of several critical factors:

  • Epitope location: For studying full-length sacsin, antibodies targeting conserved regions are preferable. When investigating truncated proteins resulting from frameshift mutations, N-terminal antibodies may be more suitable .

  • Experimental application: Different applications (Western blot, immunoprecipitation, immunohistochemistry) may require antibodies with different characteristics. For example, antibodies that work well under denatured conditions may not be effective for native proteins.

  • Specificity requirements: Validation using appropriate controls, such as SACS knockout cell lines generated by CRISPR-Cas9 technology, is essential for confirming specificity .

  • Sensitivity needs: For detecting low-abundance proteins like sacsin in patient samples, highly sensitive antibodies are necessary.

When studying ARSACS patient samples with various SACS mutations, using both N-terminal and C-terminal antibodies in parallel provides complementary information about protein expression and potential truncation products.

What validation methods ensure SACS antibody specificity?

Rigorous validation of SACS antibodies ensures reliable research outcomes:

  • Knockout controls: Testing against samples where the target protein is absent (CRISPR-Cas9-engineered sacsin knockout cell lines) provides the gold standard for confirming antibody specificity .

  • Multiple antibody concordance: Using several antibodies targeting different epitopes should yield consistent results for true sacsin detection.

  • Western blot profile analysis: Verifying that the antibody detects a protein of the expected molecular weight (~520 kDa for full-length sacsin).

  • Cross-reactivity assessment: Testing against related proteins to ensure no binding to non-target proteins occurs.

Researchers have successfully validated N-terminal antibodies using CRISPR-Cas9-generated sacsin knockout HeLa and SH-SY5Y cells, confirming specificity through the absence of signal in these negative controls .

How can researchers distinguish between full-length and truncated sacsin proteins?

Distinguishing between full-length and truncated sacsin requires specialized approaches:

  • Dual antibody approach: Using both N-terminal and C-terminal antibodies in parallel. N-terminal antibodies may detect truncated products that retain this portion of the protein, while C-terminal antibodies will only detect full-length or C-terminal fragments .

  • Molecular weight assessment: Carefully analyzing the apparent molecular weight of detected bands. Full-length sacsin appears at ~520 kDa, while truncated products will show lower molecular weights.

  • Specialized gel systems: Using gradient gels (4-12% or 4-15%) to resolve both full-length and truncated proteins in the same gel system.

What are the optimized Western blot protocols for SACS antibody applications?

Detecting sacsin by Western blot requires specialized protocols due to its large size (~520 kDa) and potentially low abundance:

  • Sample preparation:

    • Use strong lysis buffers (RIPA buffer with protease inhibitors) for complete protein extraction

    • For patient-derived fibroblasts, harvest at 80-90% confluence for optimal protein yield

  • Gel electrophoresis:

    • Use low percentage (3-5%) polyacrylamide gels or gradient gels to resolve high molecular weight proteins

    • For potential aggregates, consider mixed acrylamide-agarose gels

  • Transfer conditions:

    • Extended transfer times (overnight at low voltage) for large proteins

    • Use PVDF membranes with 0.45 μm pore size

  • Antibody incubation:

    • Primary antibodies: 1:500-1:1000 dilution, overnight at 4°C

    • Secondary antibodies: HRP-conjugated anti-mouse or anti-rabbit IgG

  • Controls:

    • Positive controls: control fibroblasts

    • Negative controls: SACS knockout cells

    • Loading controls: spectrin or other high molecular weight proteins

Protein Extraction MethodAdvantagesDisadvantagesRecommended For
RIPA BufferComplete extractionMay denature some epitopesWestern blot
NP-40 BufferPreserves protein interactionsLess efficient extractionImmunoprecipitation
8M UreaSolubilizes aggregatesHighly denaturingAggregate analysis

How can researchers investigate sacsin protein degradation pathways?

Understanding sacsin degradation requires systematic inhibition of potential pathways:

  • Proteasome inhibition studies:

    • Treat cells with MG-132 (1 μM, 3-24 hours) to inhibit proteasomal degradation

    • Monitor sacsin levels by Western blot before and after treatment

  • Autophagy inhibition:

    • Apply chloroquine (20 μM, 24 hours) to block lysosomal degradation

    • Consider combined proteasome and autophagy inhibition (0.25 μM MG-132 + 10 μM chloroquine, 18 hours)

  • Protease inhibitor treatments:

    • Test specific inhibitors: E64 (cysteine proteases), bestatin (amino peptidases), and pepstatin (aspartyl proteases)

    • Assess sacsin levels after each treatment

  • mRNA stability assessment:

    • Compare SACS mRNA levels between controls and patients to identify potential nonsense-mediated decay

    • Use RT-qPCR to quantify transcript levels in different mutation contexts

Research has shown that proteasome inhibition, autophagy inhibition, and protease inhibition do not restore mutant sacsin levels in patient fibroblasts, suggesting alternative mechanisms of protein loss that require further investigation .

What methods can detect potential sacsin protein aggregation?

Investigating potential sacsin aggregation requires specialized approaches:

  • Solubility fractionation:

    • Separate soluble and insoluble protein fractions by differential centrifugation

    • Analyze both fractions by Western blot to detect aggregated protein

  • Specialized solubilization techniques:

    • Use 8M urea buffer to solubilize potential aggregates

    • Apply mixed acrylamide-agarose gel systems to resolve high molecular weight aggregates

  • Immunofluorescence microscopy:

    • Visualize potential aggregates using SACS antibodies

    • Co-stain with markers of protein quality control (ubiquitin, p62, LC3)

  • Filter trap assay:

    • Capture large protein aggregates on cellulose acetate membranes

    • Detect trapped sacsin using specific antibodies

Despite applying these methods, studies have been unable to detect sacsin aggregates in patient fibroblasts, suggesting protein instability rather than aggregation may be the primary mechanism of protein loss in ARSACS .

How can SACS antibodies help characterize patient-specific mutations?

SACS antibodies provide crucial tools for characterizing the molecular consequences of patient-specific mutations:

  • Protein expression analysis:

    • Quantify sacsin levels in patient-derived fibroblasts with different SACS mutations

    • Compare protein expression between missense and frameshift mutations

  • Truncated protein detection:

    • Use N-terminal antibodies to identify potential truncated sacsin variants

    • Correlate truncation patterns with specific mutation types

  • Genotype-phenotype correlation:

    • Relate sacsin protein levels to clinical severity

    • Investigate whether specific mutations affect protein stability differently

  • mRNA-protein relationship:

    • Compare SACS mRNA levels with protein expression to identify post-transcriptional mechanisms

    • Determine if specific mutations affect mRNA stability versus protein stability

Research has revealed that full-length sacsin protein is dramatically reduced or completely absent in all patients with ARSACS, regardless of mutation type, which was unexpected particularly for patients carrying missense variations .

What approaches can resolve contradictory results from different SACS antibodies?

When different SACS antibodies yield contradictory results, systematic investigation is required:

  • Epitope accessibility analysis:

    • Different epitopes may be differentially accessible under various experimental conditions

    • Protein folding or post-translational modifications may mask specific epitopes

  • Truncation product investigation:

    • N-terminal antibodies may detect truncated products that C-terminal antibodies cannot

    • Compare results from both antibody types to identify potential truncated species

  • Cross-reactivity assessment:

    • Evaluate potential cross-reactivity with related proteins

    • Confirm specificity using knockout controls and peptide competition

  • Technical verification:

    • Validate that each antibody works under the specific experimental conditions used

    • Optimize protocols individually for each antibody

When studying patient-derived fibroblasts, researchers found consistent results with both C-terminal and N-terminal antibodies, showing dramatic reduction of full-length sacsin in all ARSACS patients regardless of mutation type .

How do demographic and clinical factors affect antibody response studies?

When studying antibody responses in related contexts, researchers must consider various demographic and clinical factors that may influence results:

  • Age-related variations:

    • Advanced age (≥65 years) has been associated with lower antibody responses in some studies

    • Age stratification is important for accurate interpretation of antibody data

  • Sex-based differences:

    • Male sex has been linked to lower antibody levels in certain contexts

    • Sex should be considered as a biological variable in study design and analysis

  • Clinical comorbidities:

    • Conditions like diabetes, COPD, and higher BMI may affect antibody responses

    • Stratification by comorbidities may reveal important subgroup differences

  • Previous exposure effects:

    • Prior infection can significantly influence antibody responses

    • Infection history should be documented and incorporated into analyses

These factors highlight the importance of comprehensive demographic and clinical characterization when conducting antibody-based studies, including those involving SACS antibodies in research settings.

How can researchers address the challenge of detecting low abundance sacsin protein?

Detecting low abundance sacsin protein requires specialized approaches:

  • Sample enrichment techniques:

    • Concentrate protein from larger cell populations

    • Use immunoprecipitation to enrich sacsin before Western blot analysis

  • Signal amplification methods:

    • Employ enhanced chemiluminescence systems with extended exposure times

    • Consider tyramide signal amplification for immunohistochemistry

  • Sensitive detection systems:

    • Use high-sensitivity Western blot substrates

    • Consider digital imaging systems with improved dynamic range

  • Optimized extraction protocols:

    • Test multiple lysis buffers to maximize protein recovery

    • Include phosphatase inhibitors to preserve post-translational modifications

  • Loading control selection:

    • Choose appropriate high molecular weight loading controls (e.g., spectrin)

    • Consider total protein staining methods for normalization

Despite optimization, researchers should be prepared for dramatically reduced or absent full-length sacsin signal in patient samples, as observed in multiple studies .

What strategies overcome technical challenges in SACS antibody applications?

Several strategies can address technical challenges in SACS antibody applications:

  • Large protein handling:

    • Use specialized gel systems for high molecular weight proteins

    • Optimize transfer conditions for large proteins (lower voltage, longer time)

    • Consider pulsed-field gel electrophoresis for improved resolution

  • Antibody validation approaches:

    • Generate knockout controls using CRISPR-Cas9 technology

    • Employ overexpression systems as positive controls

    • Use peptide competition to confirm specificity

  • Protocol optimization guidelines:

    • Systematically test buffer compositions, antibody dilutions, and incubation times

    • Document optimized conditions for reproducibility

    • Consider cell-type specific protocol adjustments

  • Cross-platform verification:

    • Confirm Western blot findings with immunofluorescence or flow cytometry

    • Use mass spectrometry to validate antibody-based protein identifications

These strategies ensure more reliable and reproducible results when working with challenging proteins like sacsin.

How should contradictory findings about sacsin protein levels be interpreted?

Interpreting contradictory findings about sacsin protein levels requires systematic evaluation:

  • Methodological differences:

    • Compare extraction methods, detection systems, and quantification approaches

    • Assess potential technical limitations in each study

  • Sample variation considerations:

    • Evaluate patient characteristics (mutation types, clinical severity, age)

    • Consider cell passage number and culture conditions for patient-derived cells

  • Antibody characteristic assessment:

    • Compare epitope locations, specificity profiles, and validation methods

    • Consider potential differences in sensitivity between antibodies

  • Alternative mechanisms exploration:

    • Investigate protein misfolding versus degradation

    • Consider post-translational modifications that might affect antibody recognition

  • Integrative data analysis:

    • Combine protein and mRNA data for comprehensive interpretation

    • Consider multiple time points to assess dynamic changes

Research has shown that sacsin protein is consistently reduced in ARSACS patients regardless of mutation type, suggesting fundamental disruption of protein expression or stability as a common disease mechanism .

What are the current limitations in SACS antibody research?

Current SACS antibody research faces several important limitations:

  • Antibody availability: Limited commercial options for well-validated SACS antibodies restricts research accessibility.

  • Epitope coverage: Most available antibodies target specific regions, potentially missing important variants or modified forms.

  • Sensitivity challenges: Detecting low levels of sacsin protein, particularly in patient samples, remains technically difficult.

  • Standardization issues: Lack of standardized protocols for sacsin detection complicates cross-study comparisons.

  • Mechanistic understanding gaps: The precise reasons why mutant sacsin proteins are undetectable despite stable mRNA levels in some cases remain unclear .

Despite these limitations, ongoing development of new antibodies, such as the N-terminal anti-sacsin antibody (AbN) , continues to expand the toolkit available for ARSACS research.

What future directions could enhance SACS antibody applications in research?

Several promising directions could enhance SACS antibody applications:

  • Advanced antibody engineering: Developing monoclonal antibodies against multiple sacsin epitopes would improve specificity and reproducibility.

  • Improved detection technologies: Adapting SACS antibodies for super-resolution microscopy could reveal detailed subcellular localization patterns.

  • Quantitative approaches: Establishing absolute quantification methods for sacsin would enable more precise comparison between samples.

  • Therapeutic implications: Exploring the relationship between sacsin protein levels and disease progression could inform therapeutic development.

  • Integrative multi-omics: Combining antibody-based protein detection with transcriptomics and proteomics approaches would provide comprehensive understanding of SACS mutations.

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.