MUC1 Human

Mucin-1 (CA15-3) Human
Shipped with Ice Packs
In Stock

Description

Molecular Structure of MUC1

MUC1 (Mucin 1) is a transmembrane glycoprotein encoded by the MUC1 gene. Its structure comprises two subunits formed by proteolytic cleavage:

  • N-terminal subunit (MUC1-N):

    • Contains 20-amino acid tandem repeats (VNTR) rich in serine (Ser), threonine (Thr), and proline (Pro) residues .

    • Modified by O-glycosylation, forming a heavily glycosylated extracellular domain .

    • Functions as a physical barrier and lubricant in normal epithelial cells .

  • C-terminal subunit (MUC1-C):

    • Includes a transmembrane domain and cytoplasmic tail with phosphorylation sites .

    • Mediates intracellular signaling pathways (e.g., Ras/MAPK, PI3K/Akt/mTOR) in cancer .

Table 2: MUC1 Expression in Select Cancers

Cancer TypeMUC1 Positivity RateClinical Correlation
Breast adenocarcinoma57.4% Linked to tumor size/stage
Colorectal cancer55.6% Associated with lymph node metastasis
Hepatocellular carcinoma77.1% Correlated with poor differentiation

3.1. Diagnostic Biomarker

  • Elevated serum MUC1 mRNA levels predict chemotherapy resistance in NSCLC .

  • Anti-MUC1 autoantibodies in saliva/serum show potential for early breast cancer detection .

3.2. Targeted Therapies

  • Vaccines: Phase III trials (NCT03300817) demonstrate reduced adenoma recurrence post-MUC1 vaccination .

  • Antibody-drug conjugates (ADCs): huMNC2-CAR44/22 CAR-T cells show partial responses in metastatic breast cancer .

  • Small-molecule inhibitors: Targeting MUC1-C suppresses oncogenic signaling .

Table 3: Clinical Trials Targeting MUC1

Therapy TypePhaseCancer TypeKey Outcome
MUC1 peptide vaccineIIIColorectal adenoma45.5% reduced recurrence
CAR-T (huMNC2-CAR44)IMetastatic breastStable disease in 5/12 patients
ADC (3D1-MMAE)PreclinicalTriple-negative breastTumor regression in xenografts

Research Trends and Future Directions

  • Immunology: MUC1 is the second-most prioritized cancer antigen in NIH studies .

  • Emerging areas:

    • CRISPR-engineered PD-1 knockout CAR-T cells for enhanced persistence .

    • Bibliometric analysis reveals "chimeric antigen receptor" and "T-cell" as hotspots .

Table 4: Key Research Trends (2012–2021)

Focus AreaPublicationsLeading Countries
Immunotherapy1,415USA, China, Germany
Mechanistic studies1,982USA, Japan

Challenges and Limitations

  • Tumor heterogeneity: MUC1 glycosylation patterns vary across cancers, complicating targeting .

  • Toxicity risks: Grade 3 cytokine release syndrome observed in CAR-T trials .

Product Specs

Introduction
Human Mucin-1 (CA15-3), also known as MUC1, is a glycoprotein characterized by extensive O-linked glycosylation in its extracellular domain. It comprises alpha and beta subunits that combine to form a heterodimeric complex. While the N-terminal alpha subunit plays a role in cell adhesion, the C-terminal beta subunit is involved in cell signaling. Mucins are found on the apical surface of epithelial cells in various tissues, including the stomach, lungs, intestines, eyes, and others. They contribute to the body's defense against infection by providing binding sites for pathogens through oligosaccharides present in their extracellular domain, effectively preventing pathogens from reaching the cell surface. Notably, overexpression of CA15-3 is frequently linked to cancers of the colon, breast, ovary, lung, and pancreas.
Description
Human Mucin-1 (CA15-3) possesses a molecular mass of approximately 400kDa and undergoes purification from a human carcinoma cell line.
Physical Appearance
The product appears as a clear to slightly opalescent, colorless frozen solution.
Formulation
MUC1 is provided in a buffer solution composed of 0.05M sodium phosphate (pH 7.5), 0.09% sodium azide (NaN3), and 0.15M sodium chloride (NaCl).
Stability
For optimal stability, Human MUC1 should be stored at -20°C. While it can remain stable at 4°C for up to one week, long-term storage at -20°C is recommended.
Purity
The purity of Human MUC1 is greater than 60%.
Human Virus Test
The tissue sample used in the production of Human MUC1 has undergone rigorous testing and has been found negative for HIV-1 and HIV-2 antibodies, hepatitis B surface antigen (HBsAg), hepatitis C antibodies, syphilis, and HIV/HBV/HCV by PCR.
Source

Human carcinoma cell line.

Q&A

What is the normal function of MUC1 in human epithelial cells?

MUC1 is a transmembrane glycoprotein that serves multiple critical functions in epithelial cells. It provides instructions for making mucin 1, which is part of the mucus that lubricates and protects the lining of various systems including the airways, digestive system, and reproductive system .

Unlike most mucin proteins that are secreted, MUC1 spans the cell membrane and is found specifically in epithelial cells that line body surfaces and cavities. The protein contains a mucin domain with repeated stretches of amino acids (varying from 20-100 repeats) that undergoes extensive modification through the addition of sugar molecules . These sugar chains extend outward from the protein surface, creating a protective barrier against pathogens while attracting water molecules to maintain tissue hydration and lubrication .

The cytoplasmic tail of MUC1 (MUC1-CT) extends into the cell's interior and functions in signal transduction, relaying external signals to the nucleus. Through this signaling capacity, MUC1 participates in numerous cellular processes including:

  • Cell proliferation (growth and division)

  • Cell adhesion (attachment to other cells)

  • Cell motility (movement)

  • Cell survival mechanisms

Additionally, MUC1 plays a developmental role, particularly in kidney formation, suggesting its importance beyond simple barrier functions .

What techniques are most effective for detecting MUC1 expression in human tissue samples?

Multiple complementary techniques can be employed to detect MUC1 expression in human tissues, each with specific advantages:

Immunohistochemistry (IHC): This remains the gold standard for visualizing MUC1 in tissue sections. Different monoclonal antibodies recognize specific epitopes of MUC1:

  • HMFG-1 targets the PDTR epitope

  • HMFG-2 recognizes the DTR epitope

  • SM3 binds to the PDTRP epitope

When conducting IHC for MUC1, researchers should consider that neuraminidase predigestion often enhances HMFG-1 immunoreactivity by removing sialic acid residues that may mask epitopes . The binding pattern can vary significantly between single-cell clusters and larger cell aggregates, requiring thorough sampling .

Detection challenges: Importantly, heterogeneous expression patterns are common within the same tumor types, even when grown in different experimental animals, making comprehensive sampling critical . This heterogeneity means single antibody approaches may miss significant populations of MUC1-expressing cells.

Additional detection methods include:

  • Western blotting for protein size determination

  • RT-PCR for mRNA expression analysis

  • Flow cytometry for quantitative cellular analysis

  • Mass spectrometry for detailed glycosylation pattern analysis

For optimal detection sensitivity, researchers should consider using multiple antibodies simultaneously and examining various regions within tissue samples to account for expression heterogeneity.

How does MUC1 expression differ between normal tissues and cancer?

MUC1 expression undergoes significant alterations in cancer compared to normal tissues, making it an important biomarker and therapeutic target:

Normal tissue expression:

  • Restricted to the apical surface of polarized epithelial cells

  • Highly organized glycosylation patterns with extended, branched carbohydrate structures

  • Tissue-specific expression patterns (respiratory tract, female reproductive organs, gastrointestinal tract)

  • Regulated expression levels

Cancer-associated changes:

  • Loss of polarity with expression across the entire cell surface

  • Significant overexpression (up to 10-fold in some cancers)

  • Aberrant glycosylation with truncated carbohydrate structures

  • Altered cellular localization, including cytoplasmic accumulation

  • Variable expression within tumors and between patients

In oral tongue squamous cell carcinoma (OTSCC), MUC1 expression has been detected in 79.5% of all patients and 72.7% of stage III and IV patients, with varying intensities on the membrane and in the cytoplasm . Among MUC1-positive advanced OTSCC cases, 63.6% had MUC1-positive cancer cell rates exceeding 50%, while 48.5% had rates above 80% . This heterogeneity creates significant challenges for targeted therapeutic approaches.

Experimental studies in SCID mice have further demonstrated the unpredictable nature of MUC1 expression in cancer cell lines, with considerable variation observed in the same tumors grown in different mice . This heterogeneity suggests that MUC1 gene expression in both primary tumors and metastases is not tightly controlled within particular tumor cell lines .

How is MUC1 expression regulated by hormones in human tissues?

Hormonal regulation plays a significant role in controlling MUC1 expression in specific human tissues, particularly in the reproductive system:

Progesterone regulation:
Human endometrial MUC1 is up-regulated by progesterone, particularly during hormone replacement therapy cycles . This hormonal influence helps explain the cyclical changes in MUC1 expression observed throughout the menstrual cycle, with increased expression during the secretory phase when progesterone levels are elevated .

Reproductive implications:
In the reproductive context, MUC1 serves dual functions. While generally creating a protective barrier on the endometrial surface, research in rabbits shows that during implantation, MUC1 levels first increase but are then locally reduced at the site of blastocyst attachment . This suggests a complex regulatory mechanism where the blastocyst itself may induce local changes in MUC1 expression to facilitate implantation .

Experimental approaches:
Researchers studying hormonal regulation of MUC1 commonly employ:

  • Primary endometrial cell cultures with controlled hormone exposure

  • Endometrial tissue sampling across menstrual cycle phases

  • Hormone receptor antagonist studies

  • Transgenic animal models with modified hormone response elements

  • Promoter analysis studies to identify hormone-responsive regions

Understanding these regulatory mechanisms has important implications for reproductive medicine, fertility research, and potentially for hormone-responsive cancers where MUC1 is aberrantly expressed.

What are the methodological approaches for targeting MUC1 in cancer immunotherapy?

MUC1 represents an attractive target for cancer immunotherapy due to its overexpression and aberrant modification in various cancers. Several methodological approaches have demonstrated promising results:

CAR-NK cell therapy approaches:
Recent research has developed induced pluripotent stem cell (iPSC)-derived MUC1-targeted chimeric antigen receptor natural killer (CAR-NK) cells that demonstrated significant efficacy against human oral tongue squamous cell carcinoma (OTSCC) . These engineered immune cells:

  • Exhibited significant cytotoxicity against MUC1-expressing OTSCC cells in vitro in a time- and dose-dependent manner

  • Showed substantial inhibitory effects on xenograft growth compared to both standard iPSC-derived NK cells and control groups

  • Demonstrated favorable safety profiles with no observed weight loss, severe hematological toxicity, or NK cell-mediated deaths in experimental animals

Experimental design considerations:
When developing MUC1-targeted immunotherapies, researchers must address several methodological challenges:

  • Target heterogeneity: MUC1 expression varies considerably between patients (detected in 79.5% of OTSCC cases) and within tumors themselves . This necessitates careful patient selection and potentially combination approaches targeting multiple antigens.

  • On-target/off-tumor effects: While MUC1 is overexpressed in cancers, it also appears in normal tissues including spleen, kidney, and colon . Therapeutic approaches must maximize on-target effects while minimizing off-tumor toxicity through:

    • Selective targeting of cancer-specific MUC1 glycoforms

    • Careful dose titration

    • Local administration when feasible

    • Incorporation of safety switches in cellular therapies

  • Manufacturing considerations: Production of cellular therapies like iPSC-derived CAR-NK cells involves complex processes including genetic modification, differentiation protocols, and quality control measures, contributing to high manufacturing costs .

How can researchers address MUC1 expression heterogeneity in experimental models?

MUC1 expression heterogeneity represents a significant challenge for both basic research and therapeutic development. Addressing this variability requires specialized methodological approaches:

Quantification and characterization methods:

  • Comprehensive sampling: Multiple biopsies/sections from different tumor regions

  • Single-cell analysis: Flow cytometry or single-cell RNA sequencing to quantify expression at the individual cell level

  • Digital pathology: Automated image analysis of IHC-stained sections to generate expression heat maps

  • Multiplexed detection: Simultaneous analysis of MUC1 with other biomarkers to identify correlative patterns

Experimental design considerations:
Research on human cancer cell lines grown in SCID mice has demonstrated that MUC1 expression is not tightly controlled within particular tumor cell lines, with considerable heterogeneity observed in the same tumors grown in different mice . The binding pattern varies between single-cell/small-cell clusters and larger cell aggregates .

This heterogeneity means that targeting all metastatic deposits with a single monoclonal antibody directed against the MUC1 gene product appears impossible . Researchers should therefore consider:

  • Multi-epitope targeting: Using antibody cocktails recognizing different MUC1 epitopes

  • Sequential sampling: Monitoring expression changes over time and treatment course

  • Patient-derived models: Using patient samples to maintain natural heterogeneity

  • Statistical power: Increasing sample sizes to account for variable expression

  • Threshold determination: Establishing minimum MUC1 expression levels required for therapeutic efficacy

What are the challenges in distinguishing between normal and tumor-associated MUC1 glycoforms?

The differential glycosylation of MUC1 between normal and cancerous tissues presents both opportunities and challenges for researchers:

Normal versus cancer-associated glycosylation:

  • Normal MUC1: Features extended, branched glycan structures with terminal sialylation

  • Cancer-associated MUC1: Characterized by truncated glycans and altered branching patterns

Detection methodologies:

  • Glycoform-specific antibodies:

    • SM3 antibody recognizes under-glycosylated MUC1 epitopes often exposed in cancers

    • HMFG-1 and HMFG-2 have different sensitivities to glycosylation patterns

    • Neuraminidase predigestion can enhance detection of certain epitopes by removing masking sialic acid residues

  • Lectin profiling:

    • Panels of lectins with differential binding to specific glycan structures

    • Lectin arrays for high-throughput glycosylation pattern analysis

  • Mass spectrometry:

    • Structural characterization of site-specific glycan modifications

    • Comparative glycoproteomics between normal and cancerous tissues

  • Enzymatic approaches:

    • Sequential glycosidase treatments to reveal specific structural features

    • Glycosyltransferase competition assays

Experimental challenges:

  • Technical difficulty in preserving delicate glycan structures during sample processing

  • Limited availability of glycoform-specific antibodies

  • Inter-individual variation in normal glycosylation patterns

  • Heterogeneous glycosylation within tumor cell populations

  • Ensuring specificity of targeting cancer-associated glycoforms to prevent off-target effects

For therapeutic development, researchers must carefully validate that their detection methods specifically identify tumor-associated glycoforms while sparing normal tissues to minimize potential toxicity.

How does the MUC1 cytoplasmic tail participate in cellular signaling pathways?

The cytoplasmic tail of MUC1 (MUC1-CT) plays crucial roles in cellular signaling that extend beyond MUC1's traditional barrier function:

Signaling mechanisms:
The MUC1 cytoplasmic tail relays signals from outside the cell to the nucleus, instructing cells to undergo specific changes . Through this process, MUC1 participates in:

  • Cell proliferation (growth and division)

  • Cell adhesion (attachment to other cells)

  • Cell motility (movement)

  • Cell survival mechanisms

Nuclear translocation:
The cytoplasmic tail can detach from the cell membrane and move to the nucleus through mechanisms that remain incompletely understood . Once in the nucleus, MUC1-CT is thought to help control the activity of other genes, functioning as a transcriptional co-regulator .

Methodological approaches to study MUC1-CT signaling:

  • Subcellular fractionation: Separation of nuclear and cytoplasmic fractions followed by Western blotting to detect MUC1-CT localization

  • Co-immunoprecipitation: Identification of MUC1-CT binding partners

  • Chromatin immunoprecipitation: Detection of MUC1-CT association with DNA

  • Fluorescence microscopy: Visualization of MUC1-CT translocation using fluorescently tagged constructs

  • Phospho-specific antibodies: Detection of activated forms of MUC1-CT

  • Mutagenesis studies: Creation of signaling-deficient mutants to identify key residues

Developmental connections:
Beyond its role in mature tissues, MUC1 is present in cells that form the kidneys and is thought to play a role in kidney development . This developmental function likely involves specific signaling pathways governed by the cytoplasmic domain.

What experimental approaches can evaluate MUC1-targeted therapy efficacy in preclinical models?

Developing effective MUC1-targeted therapies requires robust preclinical evaluation using various experimental approaches:

In vitro efficacy assessment:
Recent research with MUC1-targeted CAR-NK cells demonstrated significant cytotoxicity against MUC1-expressing oral tongue squamous cell carcinoma (OTSCC) cells in vitro in a time- and dose-dependent manner . Key methodological approaches include:

  • Cytotoxicity assays (MTT, LDH release, flow cytometry-based)

  • Time-course and dose-response evaluations

  • Target specificity confirmation using MUC1-positive vs. negative cell lines

  • Mechanism of action studies (apoptosis, necrosis, antibody-dependent cellular cytotoxicity)

In vivo model systems:
The same study showed that MUC1-targeted CAR-NK cells demonstrated significant inhibitory effects on xenograft growth compared to both standard NK cells and controls . Preclinical in vivo models include:

  • Cell line xenografts:

    • Human cancer cell lines expressing MUC1 implanted in immunodeficient mice

    • Allows for controlled MUC1 expression levels

    • Enables genetic manipulation of target cells

  • Patient-derived xenografts (PDX):

    • Maintains tumor heterogeneity and microenvironment

    • Better reflects clinical reality of variable MUC1 expression

    • Allows for personalized therapy testing

  • Humanized mouse models:

    • Mice with reconstituted human immune components

    • Enables evaluation of interactions with immune system

    • Critical for immunotherapy approaches

  • Transgenic models:

    • Genetically engineered to express human MUC1

    • Allows study of developmental aspects and normal tissue toxicity

Safety assessment parameters:
Comprehensive safety evaluation is critical, monitoring for:

  • Weight loss

  • Hematological toxicity (complete blood counts)

  • Organ-specific toxicity (biochemical markers, histopathology)

  • Therapy-related mortality

  • On-target/off-tumor effects on normal MUC1-expressing tissues

In the case of MUC1-targeted CAR-NK cells against OTSCC, researchers observed no weight loss, severe hematological toxicity, or NK cell-mediated deaths in experimental animals, suggesting a favorable safety profile .

How might next-generation sequencing approaches enhance MUC1 research?

Next-generation sequencing (NGS) technologies offer powerful opportunities to advance MUC1 research across multiple dimensions:

Genomic applications:

  • Characterization of MUC1 variable number tandem repeat (VNTR) polymorphisms

  • Identification of single nucleotide polymorphisms affecting MUC1 expression or function

  • Analysis of copy number variations in cancer

  • Promoter and enhancer region sequencing to identify regulatory elements

Transcriptomic approaches:

  • RNA-seq to quantify MUC1 expression across tissue types and disease states

  • Alternative splicing analysis to identify tissue-specific isoforms

  • Single-cell RNA-seq to resolve cellular heterogeneity in MUC1 expression

  • Spatial transcriptomics to map MUC1 expression within tissue architecture

Epigenomic investigations:

  • ChIP-seq to identify transcription factors regulating MUC1 expression

  • Methylation analysis of the MUC1 promoter region

  • Chromatin accessibility studies to identify regulatory elements

  • Identification of MUC1-CT binding sites in the genome

Integrated multi-omic approaches:
Combining genomic, transcriptomic, proteomic, and glycomic data can provide comprehensive insights into MUC1 biology and its role in disease. This integration could help identify:

  • Correlations between genetic variants and glycosylation patterns

  • Relationships between expression patterns and clinical outcomes

  • Novel regulatory mechanisms controlling MUC1 expression and function

These advanced sequencing approaches would significantly enhance our understanding of MUC1 heterogeneity in both normal physiology and disease states.

What potential exists for MUC1 as a diagnostic or prognostic biomarker?

MUC1's differential expression and modification in various diseases make it a promising biomarker candidate:

Diagnostic applications:
MUC1 expression has been detected in 79.5% of oral tongue squamous cell carcinoma (OTSCC) patients and 72.7% of stage III and IV patients . This high prevalence suggests potential utility as a diagnostic marker, particularly when combined with:

  • Analysis of MUC1 glycoforms specific to malignancy

  • Quantitative assessment of expression levels

  • Evaluation of cellular localization patterns

  • Correlation with other molecular markers

  • Cancer type-specific prognostic studies

  • Standardized detection and quantification methods

  • Integration with other prognostic markers

  • Consideration of specific MUC1 patterns rather than simple positive/negative classifications

Methodological approaches for biomarker development:

  • Standardized detection platforms:

    • Validated immunohistochemistry protocols

    • Quantitative PCR for mRNA expression

    • Mass spectrometry for glycoform profiling

    • Serum-based assays for circulating MUC1

  • Biomarker validation:

    • Large cohort studies with diverse patient populations

    • Longitudinal sampling to assess temporal changes

    • Correlation with treatment response and outcomes

    • Integration into existing diagnostic/prognostic algorithms

How can structural biology approaches enhance understanding of MUC1 function?

Structural biology techniques offer powerful tools to elucidate MUC1's complex functions at the molecular level:

Challenges in MUC1 structural analysis:
MUC1 presents unique challenges for structural studies due to:

  • Large size and extended conformation

  • Extensive glycosylation creating heterogeneity

  • Variable number of tandem repeats

  • Dynamic interactions between domains

Methodological approaches:

  • X-ray crystallography:

    • Determination of domain-specific structures

    • Analysis of MUC1-CT interactions with signaling partners

    • Investigation of antibody binding to specific epitopes

  • Nuclear magnetic resonance (NMR) spectroscopy:

    • Solution structure of smaller MUC1 domains

    • Studies of dynamic interactions

    • Characterization of glycan conformations

  • Cryo-electron microscopy:

    • Visualization of larger MUC1 complexes

    • Analysis of membrane integration

    • Study of conformational changes during signaling

  • Computational modeling:

    • Molecular dynamics simulations of MUC1 domains

    • Prediction of glycan-protein interactions

    • Modeling of MUC1's extended structure on cell surfaces

  • Biophysical techniques:

    • Surface plasmon resonance for binding kinetics

    • Analytical ultracentrifugation for complex formation

    • Small-angle X-ray scattering for solution conformation

These structural approaches would provide crucial insights into:

  • How glycosylation affects MUC1 conformation and function

  • The mechanism of MUC1-CT detachment and nuclear translocation

  • Structural differences between normal and cancer-associated MUC1

  • Rational design of therapeutic agents targeting specific MUC1 domains or conformations

Product Science Overview

Introduction

Mucin-1 (MUC1), also known as CA15-3, is a glycoprotein that plays a significant role in cell signaling and adhesion. It is extensively O-linked glycosylated in its extracellular domain and is known for its involvement in various human carcinomas .

Structure and Function

MUC1 is a heterodimeric complex composed of two subunits: the N-terminal alpha subunit and the C-terminal beta subunit. The alpha subunit is primarily involved in cell adhesion, while the beta subunit plays a role in cell signaling . This protein is localized to the apical membrane of epithelial cells and is crucial for maintaining the integrity of the epithelial barrier .

Clinical Significance

MUC1 is over-expressed in many types of cancer, including breast, ovarian, and pancreatic cancers. It is shed into the circulation, where it can be detected in the serum of patients using specific anti-MUC1 antibodies. The tumor marker assays CA 15-3 and CA 27.29 are commonly used to measure MUC1 levels in the blood . Elevated levels of MUC1 are often associated with the invasiveness and metastasization of cancer cells .

Diagnostic and Prognostic Value

The detection of MUC1 levels in the blood is particularly useful in monitoring the progression and treatment response of breast cancer. High levels of CA 15-3 are indicative of tumor burden and can help in assessing the effectiveness of therapy . In ovarian carcinoma, elevated MUC1 concentrations have been linked to advanced disease stages and poorer overall survival rates .

Research and Development

The Human MUCIN 1 (CA15-3) ELISA Kit is a widely used tool for detecting and quantifying MUC1 levels in serum, plasma, and cell culture media. This solid-phase sandwich Enzyme-Linked Immunosorbent Assay (ELISA) is designed to provide accurate and reliable measurements of MUC1, aiding in both research and clinical settings .

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.