CRYM Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Protein Context

The CRYM protein, encoded by the CRYM gene, functions as a thyroid hormone-binding protein and ketimine reductase. It regulates intracellular thyroid hormone (T3) signaling by sequestering T3, thereby modulating its availability for receptor binding . Structurally, CRYM shares sequence similarity with bacterial ornithine cyclodeaminases and exists in alternatively spliced isoforms .

Detection in Tissues

  • Western Blot: Detects CRYM in lysates (e.g., Jurkat cells, transfected 293T) .

  • Immunohistochemistry: Stains CRYM in human skeletal muscle (formalin-fixed) .

  • Immunoprecipitation: Pulls down CRYM from transfected lysates .

Disease-Associated Studies

  • Prostate Cancer: CRYM sequesters T3, reducing its mitogenic effects. Low CRYM expression correlates with poor prognosis and early biochemical recurrence .

  • Neurological Disorders: CRYM-expressing astrocytes in the striatum regulate perseverative behaviors, implicated in psychiatric/neurological conditions .

Thyroid Hormone Modulation

  • CRYM binds T3 with nanomolar affinity, blocking its interaction with nuclear receptors (TRα/TRβ) . Overexpression of CRYM reduces intracellular T3 levels by 18–44% in prostate cancer cells .

Astrocyte Function

  • In striatal astrocytes, CRYM knockout reduces T3 availability, altering medium spiny neuron (MSN) signaling and increasing perseverative behavior .

Antibody Development

  • CryoEM-based structural analysis enables sequence mapping of CRYM-targeting antibodies, enhancing epitope resolution .

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 orders within 1-3 business days after receiving them. Delivery times may vary depending on the method of purchase or location. Please consult your local distributors for specific delivery details.
Synonyms
CRYM antibody; CRYM_HUMAN antibody; Crystallin mu antibody; DFNA 40 antibody; DFNA40 antibody; Ketimine reductase antibody; Mu crystallin homolog antibody; Mu-crystallin homolog antibody; NADP regulated thyroid hormone binding protein antibody; NADP-regulated thyroid-hormone-binding protein antibody; OTTHUMP00000115878 antibody; THBP antibody; Thiomorpholine carboxylate dehydrogenase antibody
Target Names
CRYM
Uniprot No.

Target Background

Function
CRYM specifically catalyzes the reduction of imine bonds in brain substrates, which may include cystathionine ketimine (CysK) and lanthionine ketimine (LK). It binds thyroid hormone, a strong reversible inhibitor. CRYM is thought to be involved in regulating the free intracellular concentration of triiodothyronine and its access to nuclear receptors.
Gene References Into Functions
  • Research suggests that mu-crystallin may play a role in the development of cortical and hippocampal pyramidal cells in the early postnatal period, and in later stages, performs cell-specific functions in selected neuronal populations. PMID: 29603402
  • These findings confirm that ketimine reductase/CRYM acts as a classical imine reductase, which may explain the presence of bound pyruvate in the crystallized protein. PMID: 26173510
  • Ketimine reductase is a key enzyme in the pipecolate pathway, the primary lysine degradation pathway in the brain. PMID: 25931162
  • The expression of mu-crystallin is regulated through the AP-1 site in the promoter. PMID: 23508717
  • The purified human mu-crystallin has been confirmed to have ketimine reductase activity with a maximum specific activity similar to that of native ovine ketimine reductase. PMID: 21332720
  • Its enzymatic function has been identified as a ketimine reductase in the mammalian brain. PMID: 21332720
  • Its ketimine reductase activity is strongly inhibited by thyroid hormones. PMID: 21332720
  • This protein has been found to be differentially expressed in thalami from patients with schizophrenia. PMID: 20471030
  • This is the first report linking hyperglycemia with thyroid hormone binding protein CRYM, suggesting that the role of CRYM in diabetic complications requires further investigation. PMID: 20018174
  • CRYM has been identified as a candidate responsible for nonsyndromic deafness, through cDNA microarray analysis of human cochlear and vestibular tissues. PMID: 12471561
  • Up-regulation of mu-crystallin may play a specific and important role in the pathogenesis of facioscapulohumeral muscular dystrophy. PMID: 17451686
  • CRYM is a novel androgen-regulated gene whose expression is elevated in prostate cancer but down-regulated in castration therapy-resistant tumors. PMID: 19353593
  • Using shotgun mass spectrometry, this protein was found to be differentially expressed in the dorsolateral prefrontal cortex from patients with schizophrenia. PMID: 19165527
  • This protein has been found to be differentially expressed in the dorsolateral prefrontal cortex from patients with schizophrenia. PMID: 19110265

Show More

Hide All

Database Links

HGNC: 2418

OMIM: 123740

KEGG: hsa:1428

STRING: 9606.ENSP00000219599

UniGene: Hs.924

Involvement In Disease
Deafness, autosomal dominant, 40 (DFNA40)
Protein Families
Ornithine cyclodeaminase/mu-crystallin family
Subcellular Location
Cytoplasm.
Tissue Specificity
Expressed in neural tissue, muscle and kidney.

Q&A

What is CRYM protein and why is it significant in neuroscience research?

CRYM (Crystallin, mu) is a thyroid hormone-binding protein that shows enriched expression in the striatum and nucleus accumbens. Its significance lies in its preferential expression pattern, being markedly higher in striatal tissues compared to cortical regions across multiple species including mice, rats, and non-human primates . In non-human primates specifically, CRYM expression is notably higher in the caudate than in the putamen, suggesting region-specific functions . CRYM has gained particular attention in neurodegenerative disease research after studies revealed its expression is significantly reduced in Huntington's disease models, where mRNA levels can decrease by approximately 50% in the striatum of R6/2 mice, a widely used transgenic HD model . This reduction pattern has been confirmed through retrospective analysis of gene array datasets from both HD patients and various transgenic mouse models. The protein appears to function in the cytosol and may play a neuroprotective role, making it a valuable target for investigating striatal vulnerability in neurodegenerative conditions.

What types of CRYM antibodies are currently available for research applications?

Various CRYM antibodies have been developed for research purposes, spanning different host organisms, clonality types, and target epitopes. From the available data, researchers can access:

  • Polyclonal antibodies: Primarily raised in rabbits, these recognize multiple epitopes of the CRYM protein and show reactivity across human, mouse, and rat samples .

  • Monoclonal antibodies: Several clones are available (including 6B3 and 1G7) raised in mice, offering more specific epitope recognition .

  • Target-specific variants: Antibodies targeting specific amino acid regions of CRYM, including those recognizing AA 215-314, AA 1-314, AA 169-258, AA 215-315, and AA 154-183 .

The selection of appropriate antibody depends on the specific application, with some optimized for Western blotting, while others show better performance in immunohistochemistry, ELISA, immunofluorescence, or flow cytometry applications . For instance, some rabbit polyclonal antibodies show broad reactivity across human, mouse, and rat samples in IHC applications, while certain mouse monoclonal antibodies (like clone 1G7) offer versatility across multiple applications including Western blot, immunohistochemistry, immunofluorescence, and flow cytometry .

How should CRYM antibodies be stored and handled for optimal performance?

Proper storage and handling of CRYM antibodies are critical for maintaining their performance and extending their useful lifespan. Based on manufacturer specifications, researchers should adhere to the following guidelines:

  • Storage temperature: CRYM antibodies should be stored at -20°C for long-term preservation .

  • Format considerations: These antibodies are typically supplied in liquid format with a concentration of approximately 1.14 mg/mL .

  • Buffer composition: The standard buffer typically contains PBS with 0.05% sodium azide and 40% glycerol at pH 7.4 .

  • Safety precautions: Researchers should note that these preparations contain sodium azide, which is classified as a poisonous and hazardous substance requiring handling by trained personnel only .

  • Aliquoting recommendations: To prevent repeated freeze-thaw cycles that can degrade antibody quality, it is advisable to prepare small working aliquots upon receipt.

  • Working dilutions: For optimal results, dilutions should be application-specific, with recommended ranges of 1:50-1:200 for immunohistochemistry applications and 1:5000-1:10000 for ELISA protocols .

Careful adherence to these handling procedures helps ensure experimental reproducibility and reliable results when working with CRYM antibodies in research settings.

How can CRYM antibodies be optimally utilized in immunohistochemistry studies?

Immunohistochemistry (IHC) represents one of the primary applications for CRYM antibodies, particularly valuable for examining CRYM expression patterns in neural tissues. For optimal IHC protocols with CRYM antibodies, researchers should consider the following methodological approach:

  • Sample preparation: Tissues should be fixed with 4% paraformaldehyde and either embedded in paraffin or prepared for frozen sectioning. For CRYM detection, 10-20 μm sections are typically appropriate to visualize cellular distribution.

  • Antibody selection: Several CRYM antibodies show strong performance in IHC applications. Rabbit polyclonal antibodies have demonstrated reliable reactivity across human, mouse, and rat tissues . For higher specificity in certain applications, mouse monoclonal antibodies like clone 1G7 have also proven effective .

  • Working dilutions: For optimal staining with minimal background, a dilution range of 1:50-1:200 is recommended for most CRYM antibodies in IHC applications . Preliminary titration experiments are advisable when using a new antibody lot.

  • Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0) is generally effective for enhancing CRYM detection in fixed tissues.

  • Detection systems: Both chromogenic (DAB-based) and fluorescent secondary detection systems are compatible with CRYM antibodies. The choice depends on whether co-localization studies are planned.

In studies examining striatal vulnerability in Huntington's disease models, CRYM immunostaining has proven particularly valuable for assessing region-specific expression patterns, revealing that CRYM protein is preferentially expressed in the striatum (caudate/putamen) compared to the cerebral cortex across multiple species . This differential expression pattern provides important context for understanding the regional vulnerability observed in neurodegenerative conditions.

What are the optimal protocols for using CRYM antibodies in Western blot applications?

Western blotting represents a crucial application for quantitative assessment of CRYM protein levels, particularly when examining expression changes in disease models. For optimal Western blot protocols with CRYM antibodies, researchers should follow these methodological guidelines:

  • Sample preparation: Total protein extracts from tissues (particularly striatal and cortical regions for comparative studies) should be prepared using standard RIPA or NP-40 based lysis buffers supplemented with protease inhibitors. Protein concentration should be determined using BCA or Bradford assays.

  • Gel electrophoresis: 10-12% SDS-PAGE gels are suitable for resolving CRYM protein, which appears at an apparent molecular weight of approximately 37 kDa .

  • Transfer conditions: Standard semi-dry or wet transfer protocols using PVDF membranes are appropriate, with transfer times of 60-90 minutes at 100V (wet) or 25V for 30 minutes (semi-dry).

  • Blocking conditions: 5% non-fat dry milk or 3-5% BSA in TBST (Tris-buffered saline with 0.1% Tween-20) for 1 hour at room temperature helps minimize non-specific binding.

  • Antibody incubation: Primary CRYM antibodies should be diluted according to manufacturer recommendations, typically in the range of 1:500-1:2000 for Western blot applications. Incubation should occur overnight at 4°C with gentle agitation.

  • Detection system: HRP-conjugated secondary antibodies followed by enhanced chemiluminescence detection offer sensitive visualization of CRYM bands.

Western blot analysis has been instrumental in demonstrating that endogenous CRYM is preferentially expressed in the striatum compared to cerebral cortex across multiple species including rats, mice, and non-human primates . This technique has also revealed expression differences between caudate and putamen in non-human primates, with higher expression in the caudate region . These findings highlight the utility of Western blotting with CRYM antibodies for comparative expression studies across brain regions and species.

How can CRYM antibodies be applied in viral vector-based overexpression studies?

Lentiviral vector approaches for CRYM overexpression provide valuable insights into the protein's functional role in neuronal systems. When designing such studies with CRYM antibodies, researchers should consider the following methodological approach:

  • Vector design: Construct lentiviral vectors expressing recombinant CRYM (such as LV-Crym-HA) with appropriate epitope tags (e.g., HA-tag) to distinguish between endogenous and overexpressed protein .

  • Control vectors: Include appropriate controls such as lentiviruses expressing reporter genes (e.g., LV-GFP for tracking injection sites, LV-LacZ as a control for viral load) .

  • In vivo delivery: For CNS studies, stereotaxic injection techniques allow precise delivery to target regions such as the striatum. Co-injection of LV-Crym-HA with LV-GFP facilitates visualization of the infected region for subsequent tissue collection .

  • Expression validation: Quantitative RT-PCR can verify overexpression levels. In previous studies, LV-Crym-HA produced a 14.6-fold increase in Crym expression compared to control vectors .

  • Functional assessment: CRYM antibodies can be used to assess the effects of overexpression on disease-relevant endpoints, such as protection against mutant huntingtin toxicity.

In studies of Huntington's disease models, this approach has revealed that CRYM overexpression can reduce lesions produced by mutant huntingtin proteins. Specifically, analysis of cytochrome oxidase (COX) histochemistry showed that Crym overexpression reduced lesion volumes produced by LV-Htt171-82Q (mean COX-depleted volume of 0.236 ± 0.024 mm³ with Crym overexpression versus 0.360 ± 0.057 mm³ in controls, p < 0.05) . These findings suggest a potentially neuroprotective role for CRYM and demonstrate the utility of antibody-based detection methods in assessing the outcomes of CRYM modulation in disease models.

How can cryo-electron microscopy be integrated with CRYM antibody studies?

Cryo-electron microscopy (cryo-EM) represents an advanced structural biology technique that can be combined with antibody-based approaches to study CRYM protein complexes at high resolution. For researchers seeking to integrate these methods, the following methodological considerations are important:

  • Sample preparation: Cryo-EM requires highly purified protein samples. CRYM can be purified using standard affinity chromatography approaches, potentially using anti-CRYM antibodies in immunoaffinity purification steps .

  • Antibody-based affinity techniques: The cryo-SPIEM (cryo-Solid Phase Immune Electron Microscopy) approach combines sample purification and cryo-EM grid preparation into a single step, which is particularly valuable for low-abundance or low-yield samples like CRYM-containing complexes .

  • Complex formation: Anti-CRYM antibodies can be used to form stable CRYM:antibody complexes for structural studies. Similar to examples in the literature where antibodies form distinct macromolecular complexes with target proteins (such as the Arginase complexes described in result ), CRYM:antibody complexes could be formed for structural analysis .

  • Data collection and processing: Modern cryo-EM approaches can achieve local resolutions of 3.5 Å or better, enabling unambiguous mapping of epitopes and paratopes, which would be valuable for understanding how antibodies interact with different domains of CRYM .

  • Structural insights: Cryo-EM studies of CRYM:antibody complexes could potentially reveal orthosteric and allosteric mechanisms of interaction, similar to findings with other enzyme:antibody complexes .

This integrated approach offers several advantages, including the ability to work with native CRYM complexes directly from cell cultures without extensive purification steps . Additionally, the technique can be applied to study both high and low molecular weight complexes, those with varying symmetry properties, and those available in low concentrations . These capabilities make cryo-EM a powerful complementary approach to traditional antibody-based biochemical methods for studying CRYM structure and function.

How can CRYM antibodies help elucidate the role of CRYM in Huntington's disease pathogenesis?

CRYM antibodies serve as critical tools for investigating CRYM's potential role in Huntington's disease (HD) pathogenesis through multiple experimental approaches. Research has established that CRYM mRNA levels are significantly reduced in HD models, but the functional consequences remain under investigation . To elucidate CRYM's role in HD, researchers can implement the following methodological strategies:

  • Expression profiling: CRYM antibodies enable quantitative assessment of protein expression changes in various HD models. Western blot analysis has confirmed reduced expression in both genetic mouse models of HD (BACHD transgenic mice and Knock-in 140CAG mice) in the absence of major striatal degeneration .

  • Cellular localization studies: Immunohistochemistry with CRYM antibodies helps map expression patterns in specific neuronal populations affected in HD. Studies have shown that CRYM is localized primarily in the cytosol of striatal neurons .

  • Mechanistic investigations: Through overexpression approaches coupled with antibody detection, researchers have determined that CRYM may play a neuroprotective role. Lentiviral vector-mediated overexpression of CRYM reduced striatal lesions produced by mutant huntingtin (mHtt) as assessed by COX histochemistry (mean COX-depleted volume of 0.236 ± 0.024 mm³ with CRYM overexpression versus 0.360 ± 0.057 mm³ in controls, p < 0.05) .

  • Inclusion body assessment: CRYM antibodies can be used alongside other markers (such as Em48 for mHtt inclusions) to evaluate the impact of CRYM on disease pathology. Interestingly, CRYM overexpression appears to increase the number of Em48-containing inclusions, suggesting a complex relationship between CRYM levels and mHtt aggregation .

  • Comparative regional analysis: Given that CRYM is preferentially expressed in the striatum compared to cerebral cortex across multiple species (rats, mice, non-human primates), antibody-based detection methods help establish correlations between regional CRYM expression and differential vulnerability to HD pathology .

These approaches collectively support two possible interpretations of CRYM's role in HD: either reduced CRYM expression contributes to neuronal vulnerability (suggesting CRYM is neuroprotective), or alternatively, decreased CRYM represents a compensatory mechanism if CRYM normally contributes to striatal vulnerability . Experimental evidence using lentiviral approaches favors the neuroprotective hypothesis, as CRYM overexpression reduced mHtt toxicity in mouse models .

What considerations should be made when using CRYM antibodies for co-immunoprecipitation studies?

Co-immunoprecipitation (co-IP) with CRYM antibodies represents a powerful approach for investigating CRYM's protein interaction network and potential binding partners. When designing co-IP experiments with CRYM antibodies, researchers should consider these methodological details:

  • Antibody selection: For co-IP applications, antibodies with high specificity and affinity are essential. Polyclonal antibodies often perform well in co-IP experiments due to their recognition of multiple epitopes, potentially enhancing capture efficiency .

  • Cell/tissue lysate preparation: When preparing lysates from neural tissues, gentle lysis buffers containing 0.5-1% NP-40 or Triton X-100 with protease inhibitors help preserve protein-protein interactions. For CRYM interactions that may be dependent on thyroid hormone binding, consideration should be given to maintaining physiologically relevant hormone concentrations in buffers.

  • Pre-clearing steps: To reduce non-specific binding, pre-clearing lysates with protein A/G beads before adding the CRYM antibody is advisable.

  • Antibody immobilization: Several rabbit polyclonal anti-CRYM antibodies have been successfully used for immunoprecipitation applications . The antibody can be immobilized on protein A/G beads or directly conjugated to sepharose/magnetic beads for more efficient pull-down.

  • Validation controls: Include appropriate controls such as IgG from the same species as the CRYM antibody to identify non-specific interactions. Additionally, validation using CRYM-depleted samples (via siRNA/shRNA) helps confirm specificity.

  • Elution and analysis: After washing to remove non-specifically bound proteins, immunoprecipitated complexes can be eluted under native or denaturing conditions depending on downstream applications. Western blot analysis with antibodies against suspected interaction partners can confirm co-precipitation.

  • Mass spectrometry integration: For unbiased identification of novel CRYM interaction partners, eluted complexes can be analyzed by mass spectrometry. This approach could potentially reveal new functional roles for CRYM in thyroid hormone signaling or other cellular pathways.

Co-IP studies with CRYM antibodies could be particularly valuable for investigating whether CRYM's neuroprotective effects in HD models are mediated through direct interactions with mutant huntingtin or through modulation of thyroid hormone signaling pathways in striatal neurons.

How can researchers address common challenges when using CRYM antibodies in immunohistochemistry?

When working with CRYM antibodies in immunohistochemistry applications, researchers may encounter several challenges that require specific optimization strategies:

  • High background signal: This common issue can be addressed through:

    • More stringent blocking (extending blocking time to 2 hours or using a combination of BSA and serum from the secondary antibody host species)

    • Increasing washing steps (5-6 washes of 10 minutes each with PBST)

    • Further diluting primary antibody (testing a dilution series beyond the recommended 1:50-1:200 range)

    • Using more specific detection systems like polymer-based detection rather than avidin-biotin methods

  • Weak or absent signal: When CRYM detection is suboptimal, consider:

    • Optimizing antigen retrieval (testing multiple buffers including citrate pH 6.0, EDTA pH 8.0, or commercial retrieval solutions)

    • Extending primary antibody incubation (overnight at 4°C or up to 48 hours for challenging tissues)

    • Using signal amplification methods such as tyramide signal amplification

    • Confirming tissue fixation parameters (overfixation can mask epitopes)

  • Non-specific staining: To improve specificity:

    • Pre-absorb the antibody with purified antigen when available

    • Include appropriate negative controls (omitting primary antibody, using isotype controls)

    • Validate staining patterns with a second CRYM antibody recognizing a different epitope

    • Use tissues from CRYM knockout animals as definitive negative controls when available

  • Regional variability: Given CRYM's differential expression across brain regions, establishing appropriate exposure settings and ensuring consistent processing across samples is crucial. The notably higher expression in striatum compared to cortex provides an internal reference for validating staining patterns.

By systematically addressing these challenges, researchers can optimize CRYM antibody performance in immunohistochemistry applications, enabling reliable visualization of CRYM expression patterns across different experimental conditions and disease models.

What strategies can optimize CRYM antibody performance in Western blot applications?

Optimizing Western blot protocols for CRYM detection requires attention to several key parameters that can significantly impact sensitivity and specificity:

  • Sample preparation optimization:

    • Include phosphatase inhibitors along with protease inhibitors in lysis buffers

    • For brain tissue samples, use a Dounce homogenizer followed by sonication to ensure complete lysis

    • Heat samples at 70°C instead of 95°C to prevent potential aggregation of CRYM protein

    • Load adequate protein amounts (typically 20-40 μg for brain tissue samples)

  • Transfer optimization:

    • For the 37 kDa CRYM protein , a transfer time of 60-90 minutes at 100V in wet transfer systems typically provides efficient transfer

    • Using PVDF membranes rather than nitrocellulose may improve signal retention

    • Including methanol (10-20%) in transfer buffer can enhance transfer efficiency

  • Antibody dilution and incubation parameters:

    • Test a range of primary antibody dilutions in a small-scale optimization experiment

    • Compare overnight incubation at 4°C versus extended incubation (48-72 hours) for challenging samples

    • Consider using antibody dilution buffers containing non-animal proteins (like fish gelatin) to reduce background

  • Detection system selection:

    • For low abundance samples, enhanced chemiluminescence (ECL) plus or super signal systems offer higher sensitivity

    • Fluorescent secondary antibodies provide greater linearity for quantitative analysis

    • Digital imaging systems with extended exposure capabilities can help capture weak signals

  • Special considerations for CRYM-specific challenges:

    • When comparing CRYM levels across brain regions, include positive control samples (striatal tissue) alongside experimental samples

    • For non-human primate studies, note the expression difference between caudate and putamen regions

    • When validating antibody specificity, recombinant CRYM or lysates from cells overexpressing tagged CRYM provide useful positive controls

By implementing these optimization strategies, researchers can enhance the reliability and sensitivity of Western blot analysis for CRYM detection, enabling more accurate quantification of expression changes in various experimental models.

How might CRYM antibodies contribute to therapeutic development for neurodegenerative diseases?

CRYM antibodies have significant potential to facilitate therapeutic development for Huntington's disease and potentially other neurodegenerative conditions through several promising research avenues:

  • Target validation: CRYM antibodies provide essential tools for validating CRYM as a therapeutic target. The findings that CRYM overexpression reduces mutant huntingtin toxicity suggest that strategies to increase CRYM levels or activity could have therapeutic value. Antibodies enable quantitative assessment of such interventions.

  • Drug screening platforms: CRYM antibodies can be incorporated into high-throughput screening platforms to identify compounds that modulate CRYM expression or activity. Such screens could employ antibody-based detection methods like ELISA or high-content imaging to assess CRYM levels in response to compound libraries.

  • Pharmacodynamic biomarker development: As potential therapeutics progress toward clinical testing, CRYM antibodies could help develop pharmacodynamic biomarkers to assess target engagement. Measuring CRYM levels in accessible biospecimens might serve as a surrogate marker for central effects of treatments designed to modulate CRYM.

  • Mechanistic studies: Deeper understanding of CRYM's neuroprotective mechanisms requires identifying its interacting partners and signaling pathways. Antibody-based approaches like co-immunoprecipitation followed by mass spectrometry could reveal critical interactions that might serve as additional therapeutic targets.

  • Regional delivery approaches: Given CRYM's preferential expression in striatal regions , targeted delivery of CRYM-modulating therapies might be necessary. Antibody-based imaging could help validate the selectivity of such delivery approaches in preclinical models.

The established correlation between reduced CRYM expression and HD pathology, combined with evidence that restoring CRYM levels can attenuate mutant huntingtin toxicity , positions CRYM as a promising therapeutic target. Antibody tools will be essential for advancing these therapeutic strategies from preclinical validation through potential clinical development.

What emerging technologies might enhance CRYM antibody applications in neuroscience research?

Several cutting-edge technologies hold promise for expanding the utility of CRYM antibodies in neuroscience research:

  • Single-cell antibody-based proteomics: Emerging technologies like mass cytometry (CyTOF) and single-cell Western blotting could enable analysis of CRYM expression at the single-cell level, revealing cell-type specific expression patterns within heterogeneous brain tissues. This approach would provide unprecedented resolution of how CRYM expression varies across neuronal and glial populations in normal and disease states.

  • Advanced cryo-EM applications: The integration of antibody-based affinity techniques with cryo-electron microscopy represents a powerful approach for structural studies of CRYM . Single-step antibody-based affinity cryo-EM methods like cryo-SPIEM can combine sample purification and cryo-EM grid preparation, enabling structural studies of CRYM-containing complexes directly from cell cultures . These approaches work with both high and low molecular weight complexes and can accommodate samples available in low concentrations .

  • Spatial transcriptomics integration: Combining CRYM antibody staining with spatial transcriptomics technologies would allow correlation of CRYM protein expression with comprehensive gene expression profiles in the same tissue section, providing context for CRYM's role in broader molecular networks.

  • Antibody engineering for intrabody applications: Engineered CRYM antibody fragments (such as single-chain variable fragments) could be expressed intracellularly as "intrabodies" to modulate CRYM function or interactions in living neurons. This approach could help dissect CRYM's neuroprotective mechanisms in real-time.

  • Proximity labeling approaches: CRYM antibodies could be adapted for proximity labeling applications (such as BioID or APEX2) to identify proteins that interact with CRYM in living cells, providing insights into its functional protein complexes and thyroid hormone binding dynamics in the cellular context.

These technological advances promise to extend CRYM antibody applications beyond traditional detection methods, enabling more sophisticated investigations of CRYM's structural properties, interacting partners, and functional roles in normal and pathological contexts. Such approaches will be particularly valuable for understanding CRYM's neuroprotective mechanisms and potential as a therapeutic target in neurodegenerative diseases.

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 2024 Thebiotek. All Rights Reserved.