CYS1 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Basic Characteristics

CYS1 antibodies are immunoreagents targeting cystin 1, a 16 kDa protein encoded by the CYS1 gene. This protein localizes to primary cilia in renal ductal epithelial cells and regulates Myc proto-oncogene expression through interactions with necdin, a tumor suppressor .

Key properties of commercial CYS1 antibodies:

ParameterBoster Bio (A10081) Proteintech (24133-1-AP)
Host SpeciesRabbitRabbit
ApplicationsWB (1:1000), IHC-P (1:25)IHC (1:50–1:500), ELISA
Reactive SpeciesHuman, MouseHuman
ImmunogenSynthetic peptide (aa 128–158)Full-length CYS1 fusion protein
Storage-20°C (long-term)-20°C in 50% glycerol

Validation and Applications

CYS1 antibodies are validated across multiple platforms:

2.1. Western Blot (WB)

  • Detects endogenous CYS1 at ~16 kDa in human heart/kidney and mouse kidney lysates .

  • Optimal dilution: 1:1000 with 35 µg protein per lane .

2.2. Immunohistochemistry (IHC)

  • Localizes cystin in paraffin-embedded kidney sections .

  • Proteintech’s antibody shows reactivity in pancreatic cancer tissues with TE buffer antigen retrieval .

2.3. ELISA

  • Krishgen’s GENLISA™ (KBH2951) quantifies CYS1 in serum/plasma with monoclonal antibody pairs .

Research Findings

CYS1 antibodies have enabled critical discoveries in ARPKD pathogenesis:

3.1. ARPKD Mechanism

StudyKey Finding
Murine model (cpk mice) CYS1 mutations cause renal cysts via Myc overexpression; rescue achieved with cystin-GFP transgene.
Human case study First ARPKD patient with homozygous CYS1 splicing variant (c.318+5G>A) confirmed via allele-specific PCR.

3.2. Molecular Pathways

  • Cystin deficiency disrupts necdin-mediated Myc regulation, increasing cell proliferation in collecting ducts .

  • Rescue experiments show normalized c-MYC levels (quantified via immunoblotting) correlate with phenotypic recovery .

Implications for ARPKD Research

  • Validates cpk mice as translational models for human ARPKD .

  • Highlights Myc as a therapeutic target; c-MYC levels decrease post-cystin restoration .

  • Demonstrates GC-rich CYS1 regions complicate genetic screening, necessitating advanced sequencing .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CYS1 antibody; CYS antibody; At5g12140 antibody; MXC9.10Cysteine proteinase inhibitor 1 antibody; AtCYS-1 antibody
Target Names
CYS1
Uniprot No.

Target Background

Function
CYS1 Antibody is involved in the suppression of hypersensitive cell death triggered by either avirulent pathogens or oxidative stress.
Database Links

KEGG: ath:AT5G12140

STRING: 3702.AT5G12140.1

UniGene: At.170

Protein Families
Cystatin family, Phytocystatin subfamily
Tissue Specificity
Expressed in roots and developing siliques.

Q&A

What is the CYS1 gene and its protein product?

The CYS1 gene encodes cystin, a protein expressed in the primary apical cilia of renal ductal epithelial cells. Mutations in the CYS1 gene underlie renal cystic disease in the Cys1 (cpk/cpk) mouse model, which phenocopies human autosomal recessive polycystic kidney disease (ARPKD). Cystin has been shown to regulate Myc expression through interaction with necdin, a tumor suppressor . Understanding the protein's localization and function is essential for developing effective antibodies for research applications.

How does cystin function in normal kidney physiology?

In normal kidney physiology, cystin localizes to primary apical cilia of renal ductal epithelial cells where it appears to play a critical role in maintaining proper ciliary function. Research has demonstrated that cystin helps regulate Myc expression through protein-protein interactions. When cystin is deficient, as in the cpk mouse model, Myc expression becomes dysregulated, potentially driving the cystic kidney phenotype . This understanding provides the foundation for designing experiments where antibodies against cystin can help elucidate disease mechanisms.

What types of CYS1 antibodies are most useful for basic research?

For basic research involving cystin, investigators should consider both monoclonal and polyclonal antibodies depending on the experimental application. Monoclonal antibodies offer high specificity for particular epitopes, which is valuable for consistent detection across experiments. Polyclonal antibodies recognize multiple epitopes and may provide stronger signals for applications like immunohistochemistry. For ciliary localization studies, antibodies that can detect native conformations of cystin in fixed tissue are particularly valuable. When designing experiments to study cystin's interaction with other proteins like necdin, antibodies that don't interfere with protein binding domains are preferred .

What are the optimal methods for detecting CYS1 expression in kidney tissue?

For detecting cystin in kidney tissue, immunohistochemistry and immunofluorescence techniques have proven effective. When preparing kidney samples, proper fixation is critical - paraformaldehyde-based fixatives typically preserve ciliary structures better than alcohol-based alternatives. For immunofluorescence, confocal microscopy is recommended to visualize the precise localization within primary cilia. Co-staining with ciliary markers (such as acetylated tubulin) can help confirm proper localization. Researchers should be aware that the GC-rich nature of the CYS1 gene, particularly its first exon, can make detection challenging at the nucleic acid level . Therefore, protein-level detection via antibodies often provides more reliable results in tissue samples.

How can CYS1 antibodies be used to study ciliary dysfunction in ARPKD models?

CYS1 antibodies serve as valuable tools for investigating ciliary dysfunction in ARPKD models through several approaches:

  • Immunolocalization studies can track cystin distribution in normal versus diseased tissues

  • Co-immunoprecipitation experiments using CYS1 antibodies can identify protein interaction partners

  • Proximity ligation assays can verify in situ protein-protein interactions between cystin and suspected binding partners like necdin

  • Western blotting with CYS1 antibodies can quantify expression levels in various experimental conditions

When designing these experiments, it's important to include appropriate controls, such as tissues from the cpk mouse model as a negative control for antibody specificity. For rescue experiments, as demonstrated in studies using cystin-GFP fusion proteins, antibodies against both native cystin and GFP can help distinguish endogenous from exogenous protein expression .

What considerations are important when using CYS1 antibodies in protein-protein interaction studies?

When designing experiments to study protein-protein interactions involving cystin:

  • Validate antibody epitopes to ensure they don't interfere with known interaction domains

  • Consider using multiple antibody clones recognizing different epitopes to confirm interactions

  • For co-immunoprecipitation studies, optimize lysis conditions to preserve native protein conformations

  • Use reciprocal immunoprecipitation (pulling down with antibodies against both cystin and the interacting partner) to strengthen evidence for authentic interactions

  • Include proper negative controls, such as IgG isotype controls and samples from CYS1-deficient models

Research has shown that cystin interacts with necdin to regulate Myc expression, a finding that was uncovered through protein interaction studies. When studying such interactions, researchers should be mindful that cystin modifications might affect protein binding capabilities .

How can post-translational modifications of cystin be assessed using antibody-based techniques?

Assessing post-translational modifications (PTMs) of cystin requires specialized approaches:

  • Phosphorylation-specific antibodies can identify regulatory modifications affecting cystin function

  • For detection of other PTMs, combining immunoprecipitation with mass spectrometry provides comprehensive analysis

  • Antibodies recognizing total cystin versus modified forms can be used in parallel to determine modification ratios

When studying cysteinylation, a PTM observed in some proteins, researchers should be aware that this modification can contribute to protein heterogeneity. As demonstrated in other antibody research, cysteinylation involves capping of unpaired cysteine residues with molecular cysteine. Detection methods include chromatography and mass spectrometry to characterize subpopulations of modified proteins . Although the search results don't specifically mention cysteinylation of cystin, this analytical approach could be applied to study potential modifications of cystin in research settings.

What are the challenges in generating specific antibodies against the CYS1 gene product?

Generating specific antibodies against cystin presents several challenges:

  • The CYS1 gene's first exon is GC-rich, which can complicate antigen design and expression

  • Selection of immunogenic epitopes unique to cystin requires careful bioinformatic analysis

  • Validation of antibody specificity should include tissues from CYS1-deficient models (e.g., cpk mouse)

  • Cross-reactivity testing with related ciliary proteins is essential to ensure specificity

The GC-rich nature of the CYS1 gene has made it challenging to detect even at the genomic level, with researchers noting that "GC-rich regions can be difficult to amplify and sequence using Sanger methodology and can be missed in next-generation sequencing because low sequence complexity prevents efficient capture prior to library construction" . These same complexities may affect protein expression systems used for antibody generation.

How can researchers validate the specificity of CYS1 antibodies in experimental systems?

Comprehensive validation strategies for CYS1 antibodies include:

  • Western blot analysis comparing wild-type tissues with those from CYS1-deficient models

  • Immunoprecipitation followed by mass spectrometry to confirm target identity

  • Peptide competition assays to demonstrate epitope specificity

  • Immunohistochemistry showing expected ciliary localization in wild-type tissues and absence in knockout tissues

  • Antibody performance verification across multiple experimental techniques

For validation experiments, the cpk mouse model provides an excellent negative control, as these mice lack functional cystin. Additionally, researchers can use cell systems with CYS1 knockdown or knockout for antibody validation. The rescue experiments described in the literature, where expression of cystin-GFP fusion protein in collecting ducts of cpk mice rescued the cystic phenotype, could also serve as a validation system for antibody specificity .

How can CYS1 antibodies be used to monitor therapeutic efficacy in ARPKD models?

CYS1 antibodies provide valuable tools for monitoring therapeutic interventions in ARPKD models:

  • Immunohistochemistry with cystin antibodies can assess protein restoration following gene therapy approaches

  • Western blotting can quantify cystin expression levels after pharmacological interventions

  • Co-immunoprecipitation studies can determine if protein-protein interactions are normalized following treatment

  • Ciliary localization studies can verify proper trafficking of cystin after therapeutic intervention

In research demonstrating rescue of the cpk renal phenotype through kidney-specific expression of a cystin-GFP fusion protein, antibody-based techniques were crucial for confirming therapeutic efficacy. The studies showed that expression of the cystin-GFP fusion protein in collecting duct cells down-regulated expression of Myc in cpk kidneys, linking therapeutic intervention with molecular pathway normalization .

What is the relationship between cystin deficiency and Myc expression in ARPKD models?

Studies have established an important relationship between cystin and the Myc proto-oncogene:

  • In the cpk mouse model, cystin deficiency leads to dysregulated overexpression of Myc

  • Cystin regulates Myc expression through interaction with necdin, a tumor suppressor

  • Restoration of cystin expression in collecting duct cells down-regulates Myc expression

  • The renal cystic phenotype appears to be driven by overexpression of the Myc proto-oncogene

This relationship suggests that Myc dysregulation is a critical factor in ARPKD pathogenesis. Research has demonstrated that "the renal cystic phenotype in the mouse is driven by overexpression of the Myc proto-oncogene" . This insight provides potential therapeutic targets and emphasizes the importance of antibodies that can detect both cystin and Myc in research settings.

How do findings from the cpk mouse model translate to human ARPKD research?

The cpk mouse model shows important translational relevance to human ARPKD:

  • The phenotype of cpk mice closely resembles human ARPKD

  • Human patients with CYS1 mutations exhibit ARPKD-like phenotypes

  • Both mouse and human diseases show common molecular mechanisms involving Myc dysregulation

  • The first human case of ARPKD associated with a CYS1 mutation (c.318+5G>A) confirms the relevance of the cpk mouse as a translational model

Research has identified "the first human patient with an ARPKD phenotype due to homozygosity for a deleterious splicing variant in CYS1" . This finding validates the cpk mouse as a translationally relevant disease model. For researchers, this translational connection suggests that antibodies developed against mouse cystin might also be valuable for human studies, though species-specific validation would be necessary .

What controls should be included when using CYS1 antibodies in immunohistochemistry?

When designing immunohistochemistry experiments with CYS1 antibodies, include these essential controls:

  • Positive control: Wild-type kidney tissue with known cystin expression

  • Negative control: Tissue from cpk/cpk homozygous mice lacking functional cystin

  • Antibody controls: Isotype-matched irrelevant antibody and primary antibody omission

  • Peptide competition: Pre-incubation of antibody with immunizing peptide to confirm specificity

  • Heterozygote samples: Tissues from heterozygous (Cys1+/cpk) mice to assess dosage effects

Additionally, co-staining with markers for renal collecting ducts and ciliary structures provides contextual information about cystin localization. For human samples, tissues from individuals with confirmed CYS1 mutations can serve as negative controls, though these may be rare given the recent identification of the first human patient with a CYS1 mutation .

How can antibody microarray approaches be optimized for CYS1 detection?

For antibody microarray experiments involving CYS1 detection:

  • Include multiple antibody clones recognizing different epitopes of cystin

  • Implement the experimental strategy described in the literature for quality control:

    • Label two protein aliquots with different fluorophores (e.g., Cy3 and Cy5)

    • Conduct reciprocal labeling experiments to control for dye-specific effects

    • Apply ratio analysis to assess detection accuracy

This approach utilizes proteins prepared for regular antibody microarray experiments without requiring exogenous reference markers or absolute concentration determination of individual proteins . For CYS1-specific optimization, researchers should consider the potential impact of cystin's ciliary localization on extraction efficiency and implement extraction protocols optimized for membrane-associated proteins.

What are the best approaches for studying CYS1 mutations that affect splice sites?

Based on the identification of a human ARPKD case with a CYS1 splice site mutation (c.318+5G>A), these approaches are recommended:

  • RT-PCR analysis with primers flanking the affected exon to detect aberrant splicing products

  • Minigene constructs to evaluate splicing efficiency in vitro

  • Antibody-based detection to assess protein expression levels and localization

  • Western blotting to identify truncated or aberrant protein products

The c.318+5G>A variant affects the +5 position of the canonical donor splice site (AG/GURAGU) and is predicted to disrupt base pairing between the donor splice site of pre-mRNA and the U1snRNP of the spliceosome, leading to decreased efficiency of splice site recognition and exon skipping . Researchers should be aware that the GC-rich nature of CYS1, particularly its first exon, makes detection challenging with standard methods, potentially requiring specialized techniques for comprehensive analysis.

How do antibodies against CYS1 compare with antibodies against other ARPKD-associated proteins?

When comparing antibodies against cystin with those targeting other ARPKD-associated proteins (e.g., fibrocystin/polyductin from PKHD1):

  • Subcellular localization studies show both proteins localize to primary cilia but may occupy different microdomains

  • Expression pattern analysis reveals tissue-specific differences that may explain phenotypic variations

  • Cross-reactivity testing is essential as ciliary proteins often share structural features

  • Validation approaches should be similar, utilizing appropriate genetic models as negative controls

Since ARPKD can be caused by mutations in different genes, including PKHD1, DZIP1L, and now CYS1, comparative studies using antibodies against these different proteins can provide insights into common and distinct disease mechanisms. Research has shown that "neither TRIO-WES nor TRIO-WGS analysis revealed causative mutations in >100 cystic kidney disease genes, including PKHD1 and DZIP1L" in the patient with the CYS1 mutation , highlighting the importance of comprehensive genetic and protein analysis in ARPKD research.

What are the implications of CYS1 antibody research for understanding ciliopathies beyond ARPKD?

CYS1 antibody research has broader implications for understanding ciliopathies:

  • Comparative protein localization studies can reveal common ciliary organization principles

  • Protein interaction networks identified through co-immunoprecipitation may uncover shared signaling pathways

  • Expression pattern analysis across different ciliated tissues can explain tissue-specific manifestations

  • Functional studies may reveal mechanisms relevant to other ciliopathies

The study of cystin using specific antibodies contributes to our understanding of ciliary protein function and dysfunction, potentially informing research on other ciliopathies such as nephronophthisis, Bardet-Biedl syndrome, and Joubert syndrome. The finding that cystin deficiency affects Myc expression may also have implications for the role of Myc regulation in other ciliopathies .

How can multi-omics approaches incorporate CYS1 antibody data for comprehensive disease modeling?

Integration of CYS1 antibody data into multi-omics approaches can enhance disease modeling:

  • Correlate protein expression patterns (proteomics) with transcriptional data (transcriptomics)

  • Link protein interaction networks (interactomics) with genetic variation data (genomics)

  • Associate subcellular localization patterns with functional outcomes (phenomics)

  • Incorporate temporal and spatial expression data into developmental models

For example, researchers studying the relationship between cystin deficiency and Myc overexpression could integrate:

  • Chromatin immunoprecipitation sequencing (ChIP-seq) data to identify Myc binding sites

  • RNA-seq data to measure transcriptional changes

  • Proteomics data using CYS1 antibodies to track protein expression and modifications

  • Metabolomics data to assess downstream effects on cellular metabolism

This integrated approach would provide a more comprehensive understanding of disease mechanisms and potential therapeutic targets in ARPKD and related disorders .

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.