ASNSD1 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Structure

ASNSD1 Antibodies are affinity-purified polyclonal antibodies derived from rabbit or goat sources. They target the ASNSD1 protein, which is highly conserved across species and predominantly expressed in skeletal muscle in humans . The antibodies are validated for immunohistochemistry (IHC), Western blotting (WB), and protein array analyses. Key structural features include:

  • Immunogen Sequence: NQWQEVPASGLFRIDLKSTVISRCIILQLYPWKYISRENIIEENVNSLSQISADLPAFVSVVANEAKLYLEKPVVPLNMMLPQAALETHCSNISNVP

  • Reactivity: Human-specific, with cross-reactivity unreported in tested species

  • Conjugation: Unconjugated (SigmaAldrich, Atlas Antibodies) or unmodified (Abbeza)

Applications

ASNSD1 Antibodies are employed in diverse research contexts:

ApplicationDetailsCitations
ImmunohistochemistryDetects ASNSD1 in formalin-fixed, paraffin-embedded tissues (e.g., skeletal muscle, cancer samples). Optimal dilution: 1:50–1:200 .
Western BlottingIdentifies ASNSD1 protein bands in lysates. Recommended concentration: 0.04–0.4 µg/mL .
Protein Array AnalysisValidates specificity against 364 recombinant human protein fragments to ensure low cross-reactivity .

Validation and Specificity

ASNSD1 Antibodies undergo rigorous validation protocols:

Validation ParameterMethodOutcomeCitations
Tissue Array TestingTested on 44 normal human tissues and 20 cancer types .High specificity in skeletal muscle and adipose tissue; low/no expression in liver, kidney .
Protein Array AnalysisCross-reactivity assessed against 364 recombinant proteins .No significant cross-reactivity reported .
Subcellular LocalizationImmunofluorescence (IF) data available via Human Protein Atlas .Cytoplasmic localization in muscle cells .

Tissue Expression

ASNSD1 protein is predominantly expressed in skeletal muscle, as shown by the Human Protein Atlas :

Tissue TypeExpression LevelCellular LocalizationCitations
Skeletal MuscleHighCytoplasmic
Adipose TissueModerateCytoplasmic
LiverLow/UndetectableN/A

Cancer Relevance

ASNSD1 expression in cancer tissues has been analyzed, though no direct survival correlations are reported for the antibody. The ASNSD1 gene is associated with type 2 diabetes candidate genes, suggesting potential metabolic roles .

Experimental Models

A gene-trapped Asnsd1 knockout mouse model demonstrated severe muscle weakness, myopathy, and adipose tissue infiltration, highlighting ASNSD1’s role in muscle integrity and fat metabolism . While this study did not use ASNSD1 antibodies, it underscores the protein’s biological significance.

Significance and Future Directions

The ASNSD1 Antibody is a critical tool for elucidating the protein’s involvement in muscle physiology and metabolic disorders. Future research could explore:

  • Therapeutic Applications: Investigating ASNSD1 as a biomarker for muscle-related diseases.

  • Mechanistic Studies: Linking ASNSD1 to pathways regulating muscle-adipose tissue interactions .

  • Diagnostic Utility: Evaluating antibody performance in clinical samples for cancer or metabolic disease profiling.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship orders within 1-3 business days of receipt. Delivery times may vary based on the shipping method and destination. Please consult your local distributor for specific delivery timeframes.
Synonyms
ASND1_HUMAN antibody; ASNSD 1 antibody; Asnsd1 antibody; Asparagine synthetase domain containing 1 antibody; Asparagine synthetase domain-containing protein 1 antibody; FLJ20752 antibody; HCV NS3 transactivated protein 1 antibody; HCV NS3-transactivated protein 1 antibody; NBLA00058 antibody; NS3TP1 antibody
Target Names
ASNSD1
Uniprot No.

Q&A

What is ASNSD1 and why is it relevant for muscle research?

ASNSD1 (Asparagine Synthetase Domain Containing 1) is a protein conserved across many species with highest expression levels in skeletal muscle in humans and other species. Its relevance stems from knockout mouse studies demonstrating that ASNSD1 deficiency leads to progressive degenerative myopathy and myosteatosis (replacement of muscle with adipose tissue). This makes ASNSD1 particularly important for understanding fundamental metabolic interactions between skeletal muscle and adipose tissue. These mice exhibit severe muscle weakness and increased body fat percentage, suggesting ASNSD1's significant role in muscle metabolism and function .

Which applications are ASNSD1 antibodies validated for?

ASNSD1 antibodies are specifically validated for several key research applications including immunohistochemistry (IHC), immunocytochemistry/immunofluorescence (ICC-IF), and Western blotting (WB) . For fluorescence-linked immunosorbent assay (FLISA) applications, a dilution of 1:1000 is recommended, while appropriate dilutions for Western blot should be determined for your specific experimental conditions . These validated applications allow researchers to detect and quantify ASNSD1 in various experimental contexts, from tissue samples to cell cultures.

What are the expected histological findings when studying ASNSD1-deficient muscle tissue?

When examining ASNSD1-deficient muscle tissues, researchers should expect to observe a progressive degenerative myopathy characterized by extensive replacement of myocytes with mature adipocytes (myosteatosis). Early stages (around 14 weeks in mouse models) show scattered individual and small groups of muscle fibers with features of degeneration, including internalized nuclei, hypereosinophilia (hyaline degeneration), and loss of cross-striations. These progress to necrosis with disrupted flocculated sarcoplasm. Importantly, this myopathy demonstrates minimal myocyte regeneration, minimal interstitial inflammation, and limited fibrosis (primarily in the endomysium) . The condition predominantly affects type II (fast-twitch) muscle fibers, with type I fiber-dominant muscles being largely spared.

How does ASNSD1 deficiency phenotypically differ from other established myopathy models?

The ASNSD1-deficient mouse model presents a unique phenotype that distinguishes it from other established myopathy models such as the dystrophin-deficient mdx mouse. While both show muscle weakness, ASNSD1-deficient mice develop a progressive degenerative myopathy primarily characterized by extensive replacement of myocytes with mature adipocytes. Unlike dystrophin-deficient models, ASNSD1 deficiency results in minimal myocyte regeneration, limited interstitial inflammation, and restricted fibrosis primarily to the endomysium . Additionally, ASNSD1-deficient mice show sparing of type I fiber-dominant muscles and protection of neighboring type II fibers, suggesting complex fiber-type interactions. These mice also exhibit increased body fat percentage without liver steatosis or cardiac lipidosis, indicating a specific metabolic role for ASNSD1 in skeletal muscle rather than a generalized metabolic disorder.

What are the implications of ASNSD1's role in muscle-adipose tissue metabolic interactions?

ASNSD1-deficient mice provide valuable insights into the metabolic interactions between skeletal muscle and adipose tissue. Despite extremely elevated body fat percentage and reduced muscle mass, these mice show no signs of lipotoxicity in the liver or other organs, suggesting ASNSD1 plays a specific role in skeletal muscle metabolism rather than general lipid metabolism . This observation challenges our understanding of typical obesity-related metabolic disorders where ectopic fat accumulation generally leads to lipotoxicity. The specific transdifferentiation of muscle to adipose tissue without inflammation or significant fibrosis indicates ASNSD1 may be involved in regulating cell fate decisions in muscle tissue and controlling the balance between muscle protein synthesis and adipogenesis. This makes ASNSD1 a potential target for investigating metabolic disorders, particularly those involving muscle wasting with increased adiposity.

How might the study of ASNSD1 contribute to understanding human myopathies with myosteatosis?

ASNSD1 research may provide critical insights into human myopathies characterized by myosteatosis, which remains poorly understood in many clinical contexts. The progressive muscle degeneration with adipose replacement seen in ASNSD1-deficient mice mirrors features seen in several human disorders, including certain muscular dystrophies, age-related sarcopenia, and some metabolic myopathies . ASNSD1 was included in a set of 213 candidate genes linked to type 2 diabetes mellitus, suggesting potential connections to metabolic disorders in humans. Studying ASNSD1's function could elucidate mechanisms behind adipose infiltration in muscle tissue, identify new biomarkers for diagnosis, and potentially reveal novel therapeutic targets. The specific involvement of type II muscle fibers in ASNSD1-related pathology also has implications for understanding fiber-type-specific vulnerabilities in human myopathies.

What are the optimal protocols for immunohistochemical detection of ASNSD1 in muscle tissue?

For immunohistochemical detection of ASNSD1 in muscle tissue, researchers should follow a validated protocol starting with proper fixation. Based on established protocols for similar muscle proteins, tissues should be fixed in 10% neutral buffered formalin, embedded in paraffin, and sectioned at approximately 4 μm thickness . For epitope retrieval, Cell Conditioning 1 (CC1) for 32-48 minutes has proven effective for muscle-related proteins. Primary antibody incubation should be optimized for the specific anti-ASNSD1 antibody being used, typically at dilutions between 1:100 and 1:500. For visualization, systems such as OmniMap anti-Rb HRP with ChromoMap DAB are recommended . When analyzing ASNSD1 expression patterns, it's essential to examine both cross-sections and longitudinal sections of muscle tissue to fully appreciate structural relationships. For comparative analysis, consider double immunolabeling with markers for muscle fiber types (e.g., myosin heavy chain isoforms) to distinguish between type I and type II fibers.

What controls should be included when using ASNSD1 antibodies in experimental designs?

A robust experimental design using ASNSD1 antibodies requires several essential controls. First, include a positive control consisting of skeletal muscle tissue known to express ASNSD1 at high levels. Second, incorporate a negative control using tissue from ASNSD1 knockout models when available, or alternatively, use primary antibody omission controls . Third, implement isotype controls matched to the primary antibody to address potential non-specific binding. Fourth, when performing comparative analyses, process all samples simultaneously under identical conditions to minimize technical variation. For protein expression studies, validate antibody specificity via Western blot, confirming a single band of appropriate molecular weight. Additionally, when examining diseased tissues, include age-matched healthy controls to distinguish pathological changes from normal variations. Finally, consider using multiple antibodies targeting different epitopes of ASNSD1 to confirm specificity and rule out artifacts.

What are the recommended approaches for quantifying ASNSD1 expression in Western blot analyses?

For quantitative Western blot analysis of ASNSD1 expression, begin with optimized protein extraction from muscle tissue using a buffer containing protease inhibitors to prevent degradation. Based on research with similar muscle proteins, load 20-50 μg of total protein per lane and separate by SDS-PAGE . Transfer to a PVDF or nitrocellulose membrane and block with 5% non-fat milk or BSA. Incubate with anti-ASNSD1 antibody at the recommended dilution (starting with 1:1000 for Western blot applications) . For detection, use an appropriate HRP-conjugated secondary antibody and ECL detection system. For quantification, normalize ASNSD1 signal to a suitable housekeeping protein (e.g., GAPDH, β-actin) using densitometry software. To ensure reproducibility, run technical triplicates and validate results across biological replicates. When comparing different experimental conditions, always include samples on the same gel or implement a common reference sample across multiple gels to allow for inter-gel normalization.

How can researchers address inconsistent staining patterns when using ASNSD1 antibodies in immunohistochemistry?

Inconsistent staining patterns with ASNSD1 antibodies can result from several factors. First, check fixation conditions, as overfixation may mask epitopes while underfixation can lead to tissue degradation. Optimize antigen retrieval methods systematically, testing different buffers (citrate pH 6.0 vs. EDTA pH 9.0) and retrieval times. If background staining is problematic, increase blocking time or concentration, or try alternative blocking agents such as normal serum matching the secondary antibody species . For weak or absent staining, consider increasing primary antibody concentration or incubation time. Verify antibody specificity using Western blot analysis prior to immunohistochemistry. When examining muscle tissues specifically, be aware that ASNSD1 expression varies between fiber types, with type II fibers showing different patterns compared to type I fibers . Finally, ensure consistent tissue handling throughout your experimental protocol, as variations in processing can significantly affect staining outcomes.

What approaches can be used to distinguish between ASNSD1-related myopathy and other forms of muscle degeneration?

Distinguishing ASNSD1-related myopathy from other forms of muscle degeneration requires a multi-faceted approach. Histologically, ASNSD1-related myopathy demonstrates distinctive features including extensive adipose replacement of muscle with minimal inflammation and fibrosis, primarily affecting type II muscle fibers . This differs from dystrophin-deficient models that typically show pronounced inflammation and fibrosis. Implement a comprehensive immunohistochemical panel including ASNSD1, dystrophin, desmin, and fiber-type specific markers (fast and slow myosin heavy chains). Additionally, assess the pattern of fiber involvement using sequential sections to determine fiber-type specificity. Examine regenerative markers such as MyoD and myogenin, which show minimal activity in ASNSD1-deficient muscle compared to actively regenerating myopathies . For definitive characterization, complement histopathological findings with functional assessments (grip strength, inverted screen tests) and molecular analyses (RNA-seq, proteomics) to create a comprehensive disease signature. Finally, evaluate metabolic parameters including body composition and blood chemistries, as ASNSD1-deficient models show unique metabolic phenotypes without typical lipotoxicity in non-muscle tissues.

How should researchers interpret unexpected molecular weight variations of ASNSD1 in Western blot analyses?

When encountering unexpected molecular weight variations of ASNSD1 in Western blot analyses, researchers should systematically evaluate several possibilities. First, consider post-translational modifications such as phosphorylation, glycosylation, or ubiquitination that can alter apparent molecular weight. Second, examine potential alternative splicing, as different ASNSD1 isoforms may exist with tissue-specific expression patterns. Third, check for potential proteolytic degradation during sample preparation by adding stronger protease inhibitor cocktails or processing samples at lower temperatures. Fourth, evaluate the specificity of the antibody using knockout models or siRNA knockdown controls to confirm that detected bands represent true ASNSD1 protein . For comprehensive characterization, consider employing multiple antibodies targeting different epitopes of ASNSD1. Additionally, mass spectrometry can provide definitive identification of unexpected bands. If variations appear consistent across certain tissues or experimental conditions, this may represent biologically relevant modifications rather than technical artifacts, warranting further investigation into potential functional significance of these ASNSD1 variants in different physiological or pathological states.

What are promising research directions for elucidating ASNSD1's molecular mechanism in muscle metabolism?

Future research into ASNSD1's molecular mechanism should focus on several key areas. First, investigate ASNSD1's interactome through co-immunoprecipitation followed by mass spectrometry to identify binding partners that may reveal its functional pathways. Second, employ CRISPR-Cas9 technology to create tissue-specific and inducible knockout models to determine if ASNSD1's effects are cell-autonomous or involve cross-talk between tissues . Third, explore ASNSD1's potential role in amino acid metabolism, particularly asparagine synthesis pathways, given its domain structure. Fourth, investigate ASNSD1's involvement in muscle-specific signaling pathways regulating the balance between protein synthesis and adipogenesis, potentially focusing on mTOR, AMPK, and PPARγ pathways. Fifth, examine ASNSD1 expression and function during muscle development, regeneration, and aging to understand its temporal regulation. Finally, explore potential connections between ASNSD1 and other genes linked to metabolic disorders, particularly type 2 diabetes, as ASNSD1 was included among candidate genes for this condition . These approaches would significantly advance our understanding of how ASNSD1 functions in muscle metabolism and potentially reveal new therapeutic targets for myopathies and metabolic disorders.

What technological advances might improve detection and functional analysis of ASNSD1?

Emerging technologies could substantially enhance ASNSD1 research. Single-cell RNA sequencing combined with spatial transcriptomics would provide unprecedented insights into cell-type-specific expression patterns of ASNSD1 within heterogeneous muscle tissue, potentially revealing specialized functions in specific fiber types or satellite cells . Development of highly specific monoclonal antibodies against different epitopes of ASNSD1 would improve detection sensitivity and specificity across applications. Live-cell imaging using fluorescently tagged ASNSD1 could reveal its subcellular localization and dynamics during muscle differentiation or stress responses. CRISPR-based approaches for precise gene editing could facilitate functional domain analysis through targeted mutations. Emerging proteomics techniques such as thermal proteome profiling could identify ASNSD1's interactions with small molecules and drugs. Additionally, development of small molecule inhibitors or activators specific to ASNSD1 would provide valuable tools for acute perturbation studies. Finally, advanced imaging technologies like super-resolution microscopy combined with proximity ligation assays could visualize ASNSD1's molecular interactions in their native cellular context, providing spatial information about its function within the complex architecture of muscle tissue.

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.