YBX1 Human

Recombinant Human Y Box Binding Protein 1
Shipped with Ice Packs
In Stock

Description

Epidermal Progenitor Senescence (Source )

  • YBX1 depletion reduces cycling keratinocytes (ITGA6bri^\text{bri}/CD71bri^\text{bri}) by 60% (p < 0.01).

  • Increases senescence markers (SA-β-Gal+ cells) via CXCL1/IL-8 upregulation, rescued by CXCR2 inhibition .

AML and Translation Control (Source )

  • Proteomics identified 386 downregulated proteins post-YBX1 knockout, including RNA/DNA metabolism regulators.

  • CRISPR screens revealed YBX1 dependency on ribosomal and translation initiation genes (e.g., EEF1D) .

Viral Pathogenesis (Source )

  • YBX1 binds HCMV immediate early transcripts (e.g., UL122, UL123), enhancing polysome association by 3-fold (p < 0.001).

  • CRISPR-Cas9 YBX1 knockout reduces viral yield by 90% (p < 0.001), reversible via doxycycline-induced YBX1 reexpression .

Clinical Relevance

Cancer Prognosis (Source )

  • High YBX1 expression correlates with poor survival in breast (HR = 1.7), liver (HR = 1.9), and renal cancers (HR = 2.1) .

  • Sex-specific effects: Lung cancer females show lower YBX1 expression (Cohen’s d = 0.36) and better survival .

Therapeutic Targeting (Source )

  • YBX1 is a druggable target in adenovirus-based therapies due to its role in viral replication .

  • Clinical trials targeting YBX1 in breast and lung cancers are ongoing .

Future Directions

  • Mechanistic studies on YBX1’s role in cap-independent translation during cellular stress .

  • Development of YBX1 inhibitors for cancers with high expression (e.g., pancreatic adenocarcinoma) .

Product Specs

Introduction
As a member of the CRD-mediated complex, Y Box Binding Protein 1 (YBX1) plays a crucial role in stabilizing MYC mRNA. It is involved in various cellular processes, including the translation of mammalian selenoproteins and the regulation of pre-mRNA alternative splicing. Due to its involvement in the replication of adenovirus type 5, a widely used vector in gene therapy, YBX1 has emerged as a potential drug target for cancer treatment.
Description
Recombinantly produced in E. coli, YBX1 Human Recombinant is a single, non-glycosylated polypeptide chain comprising 347 amino acids (1-324a.a). With a molecular mass of 38.3kDa, it appears larger on SDS-PAGE due to the presence of a 23 amino acid His-tag fused to its N-terminus. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The YBX1 protein solution is provided at a concentration of 0.5mg/ml and is prepared in Phosphate Buffered Saline (pH7.4) with 20% glycerol.
Stability
For optimal storage, the solution should be kept at 4°C if it will be used within 2-4 weeks. For extended storage, it is recommended to freeze the solution at -20°C. The addition of a carrier protein, such as 0.1% HSA or BSA, is advisable for long-term storage. It is important to avoid repeated freezing and thawing of the solution.
Purity
SDS-PAGE analysis indicates that the purity of the protein is greater than 85.0%.
Synonyms
Y Box Binding Protein, NSEP1,DBPB, YB1, CCAAT-Binding Transcription Factor I Subunit A, Nuclease Sensitive Element Binding Protein, Enhancer Factor I Subunit A, Y-Box Transcription Factor, Y-Box-Binding Protein, DNA-Binding P, BP-8, CSDA2, CSDB, DBPB, MDR-NF1, NSEP-1, NSEP1, YB-1, YB1, EFI-A, YB-1, Major Histocompatibility Complex Class II Y Box-Binding Protein I.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSSEAET QQPPAAPPAA PALSAADTKP GTTGSGAGSG GPGGLTSAAP AGGDKKVIAT KVLGTVKWFN VRNGYGFINR NDTKEDVFVH QTAIKKNNPR KYLRSVGDGE TVEFDVVEGE KGAEAANVTG PGGVPVQGSK YAADRNHYRR YPRRRGPPRN YQQNYQNSES GEKNEGSESA PEGQAQQRRP YRRRRFPPYY MRRPYGRRPQ YSNPPVQGEV MEGADNQGAG EQGRPVRQNM YRGYRPRFRR GPPRQRQPRE DGNEEDKENQ GDETQGQQPP QRRYRRNFNY RRRRPENPKP QDGKETKAAD PPAENSSAPE AEQGGAE.

Q&A

What is the genomic structure and cellular localization of human YBX1?

YBX1 (Y-box binding protein-1) is encoded by a gene located on chromosome 1 (1p34) that contains eight exons and spans approximately 19 kb of genomic DNA. The gene's promoter features several E-boxes and CG-repeats that regulate YBX1 transcription, producing a 1.5 kb mRNA that encodes a 324 amino acid protein . The protein is predominantly localized in the cytoplasm where it functions as a critical regulator of mRNA translation processes . While cytoplasmic localization is most common under normal conditions, YBX1 can translocate to the nucleus during cellular stress or in cancer cells, reflecting its versatile regulatory functions across different cellular compartments.

How do researchers distinguish between the different functional domains of YBX1 protein in experimental designs?

When investigating YBX1 domains, researchers typically focus on three principal functional regions: the cold-shock domain (CSD), the N-terminal domain, and the C-terminal domain. The CSD is highly conserved evolutionarily and represents the primary nucleic acid-binding region critical for interactions with both DNA and RNA . For domain-specific studies, researchers often employ:

  • Truncation mutants (expressing specific domains)

  • Site-directed mutagenesis of key residues within domains

  • Domain-swapping experiments with related proteins

  • Fusion proteins containing specific YBX1 domains linked to reporter proteins

The cold-shock domain appears particularly crucial for YBX1's interaction with IGF2BPs and its subsequent stabilization of m6A-tagged RNA transcripts, as demonstrated in leukemia research models . Experimental designs frequently incorporate domain-specific antibodies to track localization patterns associated with different functional states.

What experimental models are most commonly used to study YBX1 function in human systems?

Current research employs multiple complementary experimental systems to investigate YBX1 function:

Model SystemApplicationsKey AdvantagesCommon Techniques
Human cancer cell linesGene function studies, protein interactionsDirectly relevant to human diseaseshRNA knockdown, CRISPR/Cas9 knockout, overexpression studies
Primary patient samplesClinical correlation studiesMost relevant to disease outcomesExpression analysis, survival correlation
Mouse modelsIn vivo leukemia studiesEvaluates systemic effectsConditional knockout models, BM transplantation
In vitro binding assaysMolecular mechanism studiesDirect measurement of interactionsRIP-seq, CLIP-seq, RNA stability assays

Researchers focused on myeloid leukemia often utilize both human cell lines and mouse models with MLL-AF9-induced leukemia, extracting lineage-negative bone marrow cells from wild-type and YBX1 conditional knockout mice . This combination allows for comprehensive analysis of both mechanistic details and physiological relevance.

How does YBX1 expression differ across cancer types, and what methodologies best capture these differences?

YBX1 expression varies significantly across different cancer types, with overexpression generally correlating with poorer outcomes in multiple solid tumors. To accurately capture these differences, researchers employ multi-layered methodological approaches:

  • Transcriptomic analysis: RNA-seq or microarray profiling to quantify mRNA expression levels across cancer types

  • Protein quantification: Western blotting, immunohistochemistry, and tissue microarrays to assess protein abundance and localization

  • Bioinformatic integration: Mining large public databases like TCGA, correlating expression with clinical parameters

  • Single-cell analysis: Examining cell-type specific expression patterns within heterogeneous tumors

Analysis of survival data has revealed that high YBX1 expression significantly correlates with poor survival outcomes in two female-only cancer sites and four mixed-sex cancer sites . For optimal cross-study comparability, researchers should standardize quantification methods and utilize appropriate housekeeping genes or reference proteins.

What are the most robust approaches for analyzing YBX1's impact on patient survival in clinical datasets?

When evaluating YBX1's clinical significance through survival analysis, researchers should implement:

  • Cox proportional hazard models incorporating biological sex and YBX1 expression as distinct covariates

  • Kaplan-Meier analyses with stratification by expression levels (typically dichotomized as high/low by median split)

  • Multivariate analyses adjusting for known prognostic factors (age, stage, grade, treatment)

  • Sex-segregated analyses to identify sex-specific associations between YBX1 expression and outcomes

In recent studies, such approaches revealed that high YBX1 expression correlated with poor survival in several cancer types, with particularly noteworthy findings in lung cancer showing differential patterns between male and female patients . The methodological rigor of controlling for biological sex as a variable has proven crucial for identifying clinically meaningful patterns that might otherwise be obscured in pooled analyses.

What techniques enable researchers to distinguish YBX1's prognostic significance from other related biomarkers?

Distinguishing YBX1's independent prognostic value requires several methodological considerations:

  • Correlation analyses: Calculate Spearman or Pearson correlations between YBX1 and other established biomarkers to identify potential confounding relationships

  • Multivariate modeling: Include both YBX1 and related biomarkers in regression models to determine independent contributions

  • Pathway analysis: Contextualize YBX1 within broader signaling networks to understand its unique position

  • Conditional knockout models: Assess the effect of YBX1 deletion in the presence of other biomarkers to establish causative relationships

Research has demonstrated that YBX1 has prognostic significance in head and neck cancer, where high mRNA expression levels correlate with poor prognosis . Additionally, in myeloid leukemia, YBX1's role appears distinct from other RNA-binding proteins, as it specifically influences m6A-tagged RNA stability through interactions with IGF2BPs .

Why should researchers incorporate biological sex as a variable when studying YBX1 expression patterns?

Incorporating biological sex as a critical biological variable in YBX1 research is increasingly essential based on emerging evidence of sex-dependent molecular mechanisms. Several compelling reasons support this approach:

  • Sex-biased signatures have been identified in 53% of clinically actionable genes within The Cancer Genome Atlas

  • Sex-specific cell death mechanisms exist (males prone to PARP-1 necrosis; females to caspase-dependent apoptosis)

  • Differences between sexes in both innate and adaptive immune functions may interact with YBX1 pathways

  • Sex-specific patterns in response to oxidative stress and sensitivity to apoptosis/autophagy have been documented

How does YBX1 expression correlate with X-chromosome genes in male versus female cancer patients?

Analysis of YBX1's relationship with X-linked genes reveals intriguing sex-specific patterns:

Methodologically, researchers investigating these relationships should:

  • Perform separate correlation analyses for male and female cohorts

  • Utilize appropriate statistical corrections for multiple testing

  • Validate findings across independent datasets

  • Investigate functional implications through pathway enrichment analyses

The observed differential correlation patterns may reflect sex-specific escape from X-chromosome inactivation or distinct regulatory mechanisms influenced by sex chromosomes.

What experimental design considerations are essential when analyzing sex-specific effects of YBX1 in cancer models?

When designing experiments to investigate sex-specific effects of YBX1, researchers should implement:

  • Balanced sex representation: Include both male and female models in appropriate numbers

  • Hormone consideration: Account for hormonal status and potential interventions (ovariectomy, castration, hormone replacement)

  • Cell line selection: Use sex-matched cell lines when testing mechanisms identified in sex-specific analyses

  • Chromosomal analysis: Consider X/Y chromosome content beyond just hormonal influences

  • Statistical powering: Ensure adequate sample sizes to detect sex-specific effects

Particularly in lung cancer, where female patients showed better survival and lower YBX1 expression compared to males , these design considerations become critical. Researchers should move beyond the common practice of sex data pooling and instead adopt sex-informed analytical approaches that can reveal important clinical and biological differences .

How does YBX1 regulate mRNA stability in cancer cells, and what methods best characterize these interactions?

YBX1 employs several mechanisms to regulate mRNA stability in cancer cells, particularly through its interaction with m6A-modified transcripts:

  • YBX1 interacts with insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) via its cold-shock domain to stabilize m6A-tagged RNA transcripts

  • This stabilization appears particularly important for specific oncogenic transcripts, including BCL2 and MYC

  • YBX1 deletion promotes mRNA decay of these m6A-tagged transcripts, contributing to apoptosis and differentiation of leukemia cells

For methodological characterization of these interactions, researchers employ:

  • RNA stability assays using actinomycin D chase experiments

  • RNA immunoprecipitation (RIP) to identify bound transcripts

  • Cross-linking immunoprecipitation (CLIP-seq) for genome-wide binding site identification

  • Luciferase reporter assays with wild-type and mutated 3'UTR sequences

These approaches have revealed that YBX1 deficiency specifically dysregulates expression of apoptosis-related genes and promotes the decay of MYC and BCL2 transcripts in an m6A-dependent manner .

What experimental approaches most effectively demonstrate YBX1's role in leukemia cell survival?

To establish YBX1's essential role in leukemia cell survival, researchers have employed multiple complementary approaches:

  • Genetic manipulation techniques:

    • shRNA-mediated knockdown with multiple targeting sequences to ensure specificity

    • CRISPR/Cas9-mediated knockout using different guide RNAs

    • Rescue experiments with shRNA-resistant YBX1 cDNA to exclude off-target effects

  • Functional assays:

    • Clonogenic assays to assess self-renewal capacity

    • Cell cycle analysis using flow cytometry

    • Apoptosis assays measuring Annexin V/PI staining

    • In vivo leukemia development using mouse models

  • Mechanistic investigations:

    • Analysis of downstream target gene expression

    • RNA stability measurements of key survival genes

    • Protein interaction studies identifying relevant binding partners

These approaches revealed that YBX1 deletion dramatically induces apoptosis and promotes differentiation in leukemia cells, while having minimal impact on normal hematopoietic function . Particularly compelling was the demonstration that YBX1 knockdown caused delayed leukemia development, which could be reversed by restoring wild-type YBX1 expression .

What techniques best characterize the molecular interactions between YBX1 and its protein binding partners?

To elucidate YBX1's interactions with protein partners such as IGF2BPs, researchers employ multiple complementary techniques:

TechniqueApplicationStrengthLimitation
Co-immunoprecipitation (Co-IP)Protein-protein interaction validationDemonstrates physical associationLimited quantitative information
Proximity ligation assay (PLA)In situ interaction visualizationSpatial resolution in intact cellsRequires specific antibodies
Isothermal titration calorimetry (ITC)Binding affinity measurementProvides thermodynamic parametersRequires purified proteins
FRET/BRETReal-time interaction monitoringLive-cell dynamicsTechnical complexity
Protein domain mappingInteraction region identificationPinpoints functional domainsLabor intensive

Research has demonstrated that YBX1 cooperates with IGF2BPs via its cold-shock domain (CSD) to stabilize m6A-tagged RNA of critical survival genes in leukemia cells . These molecular interactions form the basis for potential therapeutic targeting of YBX1 in myeloid leukemia, making accurate characterization of these interactions particularly important for drug development efforts.

How can researchers effectively validate YBX1 knockout or knockdown models to ensure specificity in cancer studies?

  • Multiple targeting strategies:

    • Use at least two independent shRNA sequences targeting different regions of YBX1

    • Employ CRISPR/Cas9 with multiple guide RNAs

    • Compare results from different genetic manipulation approaches

  • Rescue experiments:

    • Restore YBX1 expression using constructs resistant to the knockdown strategy

    • Utilize YBX1 cDNA lacking the 3'UTR for shRNAs targeting this region

    • Demonstrate restoration of phenotype with wild-type but not functionally mutated YBX1

  • Specificity controls:

    • Measure expression of highly related family members (YBX2, YBX3)

    • Perform RNA-seq to evaluate global transcriptional changes

    • Assess phenotypes in multiple cell types to detect context-dependent effects

As demonstrated in leukemia research, rescue experiments have conclusively shown that restoration of YBX1 completely rescued defects in cellular growth, clonogenic ability, cell cycle progression, and apoptosis caused by YBX1 knockdown . This methodological rigor is essential for establishing causal relationships between YBX1 and observed phenotypes.

What are the methodological challenges in analyzing m6A-mediated mechanisms of YBX1 in RNA stability?

Investigating YBX1's role in m6A-mediated RNA stability presents several technical challenges requiring specialized approaches:

  • Distinguishing direct from indirect effects:

    • Perform transcriptome-wide m6A mapping (m6A-seq or miCLIP)

    • Cross-reference with YBX1 binding sites (CLIP-seq)

    • Measure half-lives of m6A-modified vs. unmodified transcripts

  • Establishing causality:

    • Mutate specific m6A sites in target transcripts

    • Manipulate m6A writers (METTL3/14) and erasers (FTO, ALKBH5)

    • Perform rescue experiments with m6A-deficient YBX1 binding targets

  • Quantifying RNA stability accurately:

    • Use metabolic labeling (e.g., EU-seq, SLAM-seq)

    • Employ actinomycin D chase experiments with precise timepoints

    • Implement mathematical modeling to derive decay rates

Research has established that YBX1 deficiency promotes mRNA decay of MYC and BCL2 in an m6A-dependent manner , but accurately measuring these effects requires careful control of confounding variables including transcription rates, cell cycle effects, and changes in global RNA processing machinery.

What statistical approaches best capture sex-specific survival differences related to YBX1 expression?

For robust analysis of sex-specific YBX1 effects on survival outcomes, researchers should implement:

  • Cox proportional hazard modeling with interaction terms:

    • Include sex × YBX1 expression interaction to test for differential effects

    • Adjust for relevant clinical covariates (age, stage, treatment)

    • Verify proportional hazards assumptions

  • Sex-stratified analyses:

    • Perform separate analyses for male and female cohorts

    • Compare hazard ratios between sexes through formal statistical testing

    • Ensure adequate sample sizes for each stratum

  • Multiple comparison control:

    • Apply false discovery rate corrections when analyzing multiple cancer types

    • Validate findings in independent cohorts

    • Use bootstrapping to establish confidence intervals

  • Data visualization techniques:

    • Generate sex-specific Kaplan-Meier curves

    • Create violin plots to visualize expression distribution differences

    • Implement forest plots to compare effect sizes across cancer types

Analysis using these approaches revealed that high YBX1 expression was significantly associated with poor survival in multiple cancer types, with distinct patterns between sexes particularly evident in lung cancer patients .

How might YBX1 inhibition be developed as a therapeutic strategy for myeloid leukemia?

Developing YBX1 as a therapeutic target in myeloid leukemia requires systematic investigation across several research dimensions:

  • Inhibitor development strategies:

    • Design small molecule inhibitors targeting the cold-shock domain

    • Develop peptide mimetics that disrupt YBX1-IGF2BP interactions

    • Explore RNA aptamers that selectively bind YBX1

    • Investigate degrader approaches (PROTACs) directed at YBX1

  • Therapeutic window assessment:

    • Compare effects on leukemic versus normal hematopoietic cells

    • Identify synergistic combinations with standard therapies

    • Determine minimal effective dosing regimens

  • Resistance mechanisms:

    • Characterize potential compensatory pathways

    • Investigate alternate m6A readers that might substitute for YBX1

    • Monitor for YBX1 mutations that prevent inhibitor binding

Current research demonstrates that YBX1 is selectively required for myeloid leukemia cell survival while being dispensable for normal hematopoiesis , providing a strong biological rationale for therapeutic targeting. The involvement of YBX1 in stabilizing critical oncogenes like BCL2 and MYC through m6A-dependent mechanisms offers multiple intervention points for drug development .

What experimental approaches could better clarify the mechanistic basis of sex-specific YBX1 effects in cancer?

Elucidating the mechanistic underpinnings of sex-specific YBX1 effects requires innovative experimental approaches:

  • Integrated multi-omics studies:

    • Compare male and female tumors using proteomics, transcriptomics, and epigenomics

    • Identify sex-specific YBX1 binding partners and regulatory networks

    • Map sex-specific post-translational modifications of YBX1

  • Chromosome-focused investigations:

    • Examine YBX1 interactions with sex chromosome genes

    • Study YBX1 in the context of X-chromosome inactivation

    • Analyze sex-specific enhancer-promoter interactions affecting YBX1

  • Hormone response elements:

    • Map sex hormone receptor binding sites near YBX1 and its targets

    • Perform hormone manipulation studies and measure YBX1 activity

    • Develop hormone-responsive reporter systems for YBX1 function

  • Clinical validation studies:

    • Design sex-stratified clinical trials for YBX1-targeting therapies

    • Collect and analyze sex-specific biomarkers of YBX1 activity

    • Develop companion diagnostics accounting for sex differences

Research has already identified that YBX1 expression correlates differently with X-linked genes in male versus female bladder cancer patients , suggesting potentially distinct regulatory mechanisms that warrant further investigation for therapeutic implications.

What next-generation sequencing approaches would most advance our understanding of YBX1's genome-wide regulatory functions?

Several cutting-edge sequencing methodologies could significantly advance YBX1 research:

  • Single-cell multi-omics:

    • scRNA-seq combined with YBX1 ChIP-seq to correlate binding with expression

    • Single-cell ATAC-seq to examine chromatin accessibility changes upon YBX1 manipulation

    • Spatial transcriptomics to map YBX1 activity in heterogeneous tumor microenvironments

  • RNA-protein interaction mapping:

    • Enhanced CLIP-seq methods with improved crosslinking efficiency

    • eCLIP combined with m6A-seq to identify YBX1 binding at m6A sites

    • RNA Bind-n-Seq to determine YBX1 sequence preferences in different contexts

  • Long-read sequencing applications:

    • Nanopore direct RNA sequencing to identify YBX1-dependent RNA modifications

    • PacBio sequencing to characterize YBX1's impact on alternative splicing

    • Long-read approaches to identify YBX1-regulated transcript isoforms

  • Integrative genomics:

    • Hi-C combined with YBX1 ChIP-seq to examine 3D genome organization

    • Ribosome profiling to assess YBX1's impact on translation efficiency

    • CRISPR screens combined with YBX1 manipulation to identify synthetic lethal interactions

These approaches could provide unprecedented insights into how YBX1 coordinates gene expression programs that drive cancer progression and create sex-specific vulnerabilities, potentially opening new avenues for precision oncology approaches .

Product Science Overview

Introduction

Recombinant Human Y Box Binding Protein 1 (YBX1) is a multifunctional protein that plays a crucial role in various cellular processes. It is a member of the Cold Shock Domain protein family and is known for its ability to bind both DNA and RNA. YBX1 is involved in transcription regulation, RNA stabilization, mRNA splicing, DNA repair, and translational repression .

Structure

YBX1 consists of 324 amino acids and has a molecular mass of approximately 38.3 kDa . The protein contains a Cold Shock Domain, which is responsible for its nucleic acid-binding properties. This domain allows YBX1 to interact with specific RNA and DNA sequences, thereby influencing various cellular functions .

Functions
  1. RNA Binding and Stabilization: YBX1 preferentially binds to the 5’-[CU]CUGCG-3’ RNA motif and recognizes mRNA transcripts modified by C5-methylcytosine (m5C). This binding promotes mRNA stabilization by preventing mRNA decay .
  2. Transcription Regulation: YBX1 can bind to promoters containing a Y-box (5’-CTGATTGGCCAA-3’), such as those of the MDR1 and HLA class II genes. This binding regulates the transcription of these genes .
  3. DNA Repair: YBX1 promotes the separation of DNA strands that contain mismatches or are modified by cisplatin. It also has endonucleolytic activity, introducing nicks or breaks into double-stranded DNA, suggesting a role in DNA repair .
  4. Alternative Splicing: YBX1 is involved in pre-mRNA alternative splicing regulation by binding to splice sites in pre-mRNA and regulating splice site selection .
  5. Extracellular Functions: The secreted form of YBX1 acts as an extracellular mitogen, stimulating cell migration and proliferation .
Role in Cancer

YBX1 is overexpressed in various human cancers and is recognized as an oncogenic gene associated with poor prognosis. Its functional diversity arises from its capacity to interact with a broad range of DNA and RNA molecules, implicating its involvement in diverse cellular processes. YBX1’s role in cancer includes promoting tumor progression, influencing the tumor microenvironment, and contributing to drug resistance .

Therapeutic Potential

Given its significant role in cancer, YBX1 is considered a potential target for cancer therapy. Ongoing clinical trials are exploring YBX1 as a target in breast cancer and lung cancer. The emerging role of YBX1 in neural input is also being investigated, with high levels of YBX1 associated with nerve cancer and 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 2025 TheBiotek. All Rights Reserved.