CEACAM1 Human

Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Immune Regulation

  • T Cell Inhibition: L-isoform ITIMs recruit SHP-1/2 phosphatases to dampen TCR signaling .

  • Neutrophil Modulation: Regulates CEACAM3-mediated bacterial phagocytosis via Lyn/Hck-dependent ITIM phosphorylation .

  • Monocyte Survival: Suppresses apoptosis via PI3K/Akt/Bcl-2 pathways .

Tumor Biology

  • Colorectal Cancer: siRNA knockdown reduces proliferation by 61% and increases apoptosis (1.23% vs. 0.197% in controls) .

  • Melanoma: Elevated CEACAM1 on PD1+ NK/T cells correlates with immunotherapy resistance .

Organ Protection

  • Liver Injury: Hypoxia-induced CEACAM1-S isoform mitigates ischemia-reperfusion injury by inhibiting ASK1/p38 signaling .

Cancer Prognosis

Cancer TypeCEACAM1 RoleClinical AssociationSource
ColorectalPro-proliferativesiRNA silencing reduces tumor growth
MelanomaImmune checkpoint co-expression (PD1)Resistance to anti-PD1 therapy
HepatocellularCEACAM1-S:HIF1A ratioPredicts liver transplant success

Therapeutic Targeting

  • Morpholinos: Nucleic acid blockers increase CEACAM1-S, protecting hepatocytes from hypoxia .

  • Antibody Therapy: Anti-CEACAM1 agents (e.g., 26H7) are under investigation for melanoma .

Pathogen Interactions

CEACAM1 serves as a receptor for Neisseria gonorrhoeae Opa proteins. Humanized transgenic mice demonstrate neutrophil binding analogous to human infections .

Future Directions

  • Isoform-Specific Therapies: Targeting CEACAM1-S in liver transplants .

  • Combination Immunotherapy: Dual CEACAM1/PD1 blockade for resistant cancers .

Product Specs

Introduction
Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (CEACAM1) is a member of the carcinoembryonic antigen (CEA) gene family, which belongs to the immunoglobulin superfamily. As a cell adhesion protein, CEACAM1 facilitates calcium-independent homophilic cell adhesion. This surface glycoprotein is expressed on various cell types, including blood cells, epithelial cells, and vascular cells. CEACAM1 acts as a coinhibitory receptor in immune responses and plays a role in angiogenesis activation.
Description
Produced in Sf9 Baculovirus cells, CEACAM1 is a single, glycosylated polypeptide chain consisting of 405 amino acids (35-428 a.a.). It has a molecular mass of 44.6 kDa. However, on SDS-PAGE, the molecular size appears approximately between 40-70 kDa. The protein is expressed with a 6 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
The CEACAM1 protein solution (0.25mg/ml) is supplied in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), store the vial at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms
Carcinoembryonic Antigen Related Cell Adhesion Molecule 1, Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 (Biliary Glycoprotein), CD66a Antigen, BGP1, BGP, Biliary Glycoprotein 1, Antigen CD66, BGP-1, BGPI.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ADPEFQLTTE SMPFNVAEGK EVLLLVHNLP QQLFGYSWYK GERVDGNRQI VGYAIGTQQA TPGPANSGRE TIYPNASLLI QNVTQNDTGF YTLQVIKSDL VNEEATGQFH VYPELPKPSI SSNNSNPVED KDAVAFTCEP ETQDTTYLWW INNQSLPVSP RLQLSNGNRT LTLLSVTRND TGPYECEIQN PVSANRSDPV TLNVTYGPDT PTISPSDTYY RPGANLSLSC YAASNPPAQY SWLINGTFQQ STQELFIPNI TVNNSGSYTC HANNSVTGCN RTTVKTIIVT ELSPVVAKPQ IKASKTTVTG DKDSVNLTCS TNDTGISIRW FFKNQSLPSS ERMKLSQGNT TLSINPVKRE DAGTYWCEVF NPISKNQSDP IMLNVNYNAL PQENGLSPGH HHHHH.

Q&A

What is the molecular structure of human CEACAM1?

Human CEACAM1 is a single-pass type I transmembrane protein expressed as 12 alternatively spliced isoforms. All isoforms contain an N-terminal V set fold of the immunoglobulin superfamily (IgV) ectodomain followed by up to three type 2 constant immunoglobulin (IgC2) ectodomains (A1, B, A2), a transmembrane sequence, and a cytoplasmic domain . The cytoplasmic domain exists in either a long (L) variant containing immunoreceptor tyrosine-based inhibitory motifs (ITIMs) or a short (S) variant devoid of ITIMs, depending on the inclusion or absence of exon 7 . Each isoform is designated by the number of extracellular domains and the presence of an L or S cytoplasmic tail (e.g., CEACAM1-4L, CEACAM1-1S) .

How do CEACAM1 expression patterns differ between normal and disease states?

CEACAM1 is typically absent or expressed at low levels on healthy circulating immune cells but shows increased expression on immune cells in the peripheral blood and tumors of patients with diseases such as melanoma . In thyroid tissue, CEACAM1 is not appreciably expressed in normal tissue and rarely present in benign tumors, but becomes highly upregulated in thyroid carcinomas . The expression pattern shift is particularly notable in treatment-resistant melanoma, where distinct immune cell populations within the tumor microenvironment exhibit high CEACAM1 levels, including tumor-infiltrating CD8+ T cells . This differential expression makes CEACAM1 a valuable marker for disease progression and potential therapeutic target.

What techniques are most effective for distinguishing between CEACAM1 isoforms in research samples?

When researching CEACAM1 isoforms, researchers should consider:

  • Antibody selection: Use antibodies that can distinguish human CEACAM1 from other highly related CEACAM family members. Specialized antibodies labeled with isotopes like 159Tb have been successfully used in high-dimensional analyses .

  • Splicing analysis: RT-PCR with isoform-specific primers can identify the presence of alternatively spliced variants, particularly distinguishing between those with long (L) or short (S) cytoplasmic domains.

  • High-dimensional cytometry: Cytometry by time-of-flight (CyTOF) provides superior resolution for analyzing CEACAM1 expression across multiple immune cell populations simultaneously. This method allows for comprehensive immune cell atlasing and has been used to create detailed inventories of CEACAM1 expression in melanoma patients .

  • Western blotting with isoform discrimination: Using antibodies that target specific domains can differentiate between membrane-bound isoforms and secreted variants such as CEACAM1-3 and CEACAM1-3C2 .

How does CEACAM1 modulate immune cell function?

CEACAM1 functions as an inhibitory receptor across multiple immune cell types through several mechanisms:

  • T cells: CEACAM1 acts as a molecular rheostat for T cell receptor signaling, influencing the balance between activation and tolerance. The absence of CEACAM1 on T cells has been shown to abrogate the development of tolerance in vivo . CEACAM1-S isoforms promote the development of regulatory T cells, while both S and L isoforms participate in different aspects of tolerance .

  • Natural Killer (NK) cells: Homophilic interactions between CEACAM1 expressed on NK cells and tumors inhibit NK cell-mediated killing, independent of MHC class I recognition . This mechanism can be therapeutically targeted with antibody fragments like DIATHIS1 to enhance NK cell cytotoxicity against CEACAM1-expressing melanoma cells .

  • Neutrophils: CEACAM1 is rapidly trafficked to the membrane from intracellular granule stores during neutrophil activation where it functions as a cellular receptor for bacterial pathogens and as a negative regulator of neutrophil function . It mediates SHP-1-dependent inhibition of the granulocyte colony-stimulating factor receptor (G-CSFR)-signal transducer and activator of transcription (Stat3) pathway, affecting granulopoiesis .

  • Monocytes: CEACAM1 promotes monocyte survival by suppressing both intrinsic (mitochondrial-mediated) and extrinsic (CD95 ligand-induced) apoptosis through upregulation of Bcl-2 mediated by PI3K and Akt-dependent signaling pathways .

What are the key signaling pathways downstream of CEACAM1 activation?

CEACAM1 signaling primarily occurs through its ITIM domains in the long (L) cytoplasmic tail variants, with several key pathways:

  • SHP-1/SHP-2 recruitment: Upon ligand binding, the ITIMs in CEACAM1-L become phosphorylated by Src family kinases (such as Lyn and Hck), recruiting protein tyrosine phosphatases SHP-1 and SHP-2 .

  • Inhibition of activating receptors: CEACAM1 negatively regulates signaling from activating receptors such as T cell receptors, B cell receptors, and cytokine receptors like G-CSFR through SHP-1-dependent dephosphorylation events .

  • Cell survival signaling: In monocytes, CEACAM1 activates PI3K and Akt-dependent signaling pathways, leading to upregulation of Bcl-2 and downregulation of caspase 3 activation, promoting cell survival .

  • Self-regulation mechanism: During advanced apoptosis, the inhibitory function of CEACAM1 is mitigated when the long cytoplasmic tail undergoes caspase 3-mediated proteolytic cleavage at a membrane proximal cytoplasmic DQRD motif, rendering it devoid of its ITIM domains .

How does CEACAM1 contribute to T cell exhaustion in cancer and chronic infections?

CEACAM1 plays a significant role in T cell exhaustion through the following mechanisms:

  • Co-inhibitory receptor function: CEACAM1 functions alongside other inhibitory receptors (CTLA-4, PD-1, LAG-3, TIM-3, TGIT, 2B4) to transduce inhibitory signals upon interactions with their ligands on target cells .

  • Progressive dysfunction: Unlike T cell anergy (which results from T cell priming without costimulatory signals), exhausted T cells arise from gradual modulation of T cell activity in the presence of continuous TCR stimulation during chronic conditions like persistent infections, autoimmunity, or malignancy .

  • Tumor immune evasion: In malignancies, CEACAM1-mediated exhaustion of tumor-specific T cells impedes the immune system's ability to eradicate tumors, allowing tumor escape .

  • Chronic viral infections: Exhausted T cells expressing CEACAM1 have been observed in response to persistent viral infections including HIV, HBV, HCV, and HTLV1 .

  • Balance in autoimmunity: Interestingly, mice with deficient T cell exhaustion develop severe spontaneous or induced autoimmune diseases, suggesting an important regulatory role for this process .

How does CEACAM1 expression correlate with cancer progression and treatment response?

CEACAM1 expression correlates with cancer progression and treatment response in several important ways:

  • Melanoma prognosis: De novo expression of CEACAM1 is strongly associated with reduced disease-free survival in patients with metastatic melanoma .

  • Treatment resistance: CEACAM1 levels are highest in treatment-resistant disease, particularly on various immune cell populations within the tumor microenvironment, including tumor-infiltrating CD8+ T cells .

  • Thyroid cancer: While not appreciably expressed in normal human thyroid tissue and rarely present in benign tumors, CEACAM1 becomes highly upregulated in thyroid carcinomas .

  • Immune evasion mechanism: The inhibitory function of CEACAM1 expressed on both tumor cells and immune cells (particularly NK cells) promotes tumor immune evasion, potentially hindering the efficacy of immunotherapeutic treatments .

  • Isoform relevance: Research indicates that melanoma cell lines with higher relative levels of CEACAM1-3L and CEACAM1-3S splice variants show greater response to treatments targeting CEACAM1, suggesting isoform-specific roles in treatment susceptibility .

What methodologies are most effective for studying CEACAM1 in the context of cancer immunotherapy?

Researchers investigating CEACAM1 in cancer immunotherapy should consider these methodological approaches:

  • High-dimensional immune cell profiling: Cytometry by time-of-flight (CyTOF) with antibodies that distinguish human CEACAM1 from other CEACAM family members enables comprehensive mapping of CEACAM1 expression across immune cell populations in the tumor microenvironment and peripheral blood .

  • Co-expression analysis: Analyze co-expression patterns of CEACAM1 with other inhibitory receptors (like PD1 and PD-L1) across immune cell subsets to identify potential synergistic therapeutic targets. This approach has revealed that the majority of circulating PD1-positive NK cells, innate T cells, B cells, monocytic cells, dendritic cells, and CD4+ T cells in treatment-resistant melanoma co-express CEACAM1 .

  • Functional antibody screening: In vitro experimental models using human antibody fragments (like scFv DIATHIS1) to block CEACAM1 homophilic interactions can assess enhancement of NK cell-mediated cytotoxicity against CEACAM1-expressing melanoma cells .

  • Degranulation assays: Measure activation and degranulation capacity of NK cells following pretreatment of melanoma cells with CEACAM1-targeting agents to evaluate immunotherapeutic potential .

  • Isoform expression analysis: Quantify relative expression levels of different CEACAM1 splice variants (particularly CEACAM1-3L and CEACAM1-3S) in tumor samples to predict potential therapeutic response .

How can CEACAM1-targeted therapies enhance existing immunotherapy approaches?

CEACAM1-targeted therapies show promise for enhancing existing immunotherapies through several mechanisms:

  • Combination with checkpoint inhibitors: Given that CEACAM1 functions as an inhibitory receptor alongside other checkpoint molecules like PD1, agents targeting CEACAM1 may represent appropriate partners for PD1-related pathway therapies .

  • Overcoming treatment resistance: The high expression of CEACAM1 in treatment-resistant disease suggests that targeting this molecule could potentially resensitize tumors to existing immunotherapies .

  • Enhancing NK cell function: Human antibody fragments like DIATHIS1 that target CEACAM1 have been shown to enhance the lytic machinery of NK cells against CEACAM1-expressing melanoma cells . In experimental models, coincubation of scFv DIATHIS1 with CEACAM1 melanoma cells and NK-92 cell line significantly increased cell-mediated cytotoxicity .

  • Promoting immune cell activation: Pretreatment of melanoma cells with CEACAM1-targeting agents promotes the activation and degranulation capacity of NK cells, potentially enhancing their anti-tumor activity .

  • Isoform-specific targeting: Targeting specific CEACAM1 isoforms (particularly CEACAM1-3L and CEACAM1-3S) might provide more precise therapeutic approaches, as melanoma cells with higher expression of these variants show greater response to CEACAM1-targeting treatments .

How do bacterial pathogens exploit CEACAM1 for infection and immune evasion?

Bacterial pathogens have evolved sophisticated mechanisms to exploit CEACAM1:

  • Streptococcus pyogenes interaction: The R28 protein of S. pyogenes contains an IgI3 domain that specifically targets human CEACAM1 to promote events favoring infection, including enhancing bacterial adhesion to epithelial barriers, interfering with wound healing, subverting immune responses, and promoting bacterial replication in blood .

  • Species specificity: The interaction between bacterial proteins and CEACAM1 often shows species specificity. For example, R28-IgI3 binds to human but not macaque CEACAM1 due to critical amino acid differences (particularly residue F29 in human CEACAM1) .

  • Multiple barrier disruption mechanism: While pathogens typically employ distinct molecular interactions to overcome each host defense mechanism, a single interaction with CEACAM1 can allow pathogens to overcome multiple host defense barriers simultaneously .

  • Neutrophil function modulation: During Neisseria gonorrhoeae infection, CEACAM3 (an ITAM-containing CEACAM family member) is first engaged by bacterial Opa proteins, triggering neutrophil activation. CEACAM1 is subsequently upregulated and accessible for Opa binding, leading to ITIM phosphorylation that negatively regulates neutrophil function .

What experimental approaches best characterize CEACAM1-pathogen interactions?

Researchers studying CEACAM1-pathogen interactions should consider these methodological approaches:

  • Structural analysis and molecular docking: Simulated docking of bacterial proteins onto wildtype and mutated CEACAM1-N domain structures can predict binding interactions and species specificity. This approach successfully predicted that R28-IgI3 binds to human but not macaque CEACAM1 .

  • Site-directed mutagenesis: Introducing specific mutations (like F29I in human CEACAM1 or I29F in macaque CEACAM1) can confirm critical residues for binding specificity. Analysis showed that F29 in human CEACAM1 is a critical determinant of human-specificity through interaction with residues K45 and I52 on R28-IgI3 .

  • Binding energy calculations: In silico analysis can quantify the effects of mutations on binding energy, providing insights into the molecular basis of species specificity .

  • Functional assays: Experiments measuring bacterial adhesion, epithelial barrier integrity, wound healing, immune cell function, and bacterial replication in the presence of CEACAM1-targeting agents can elucidate the functional consequences of CEACAM1-pathogen interactions .

  • Species-comparative approaches: Comparing CEACAM1 from different species (like human and macaque) provides insights into evolutionary adaptation and host specificity of pathogens .

What are the key methodological considerations for high-dimensional mapping of CEACAM1 expression?

When performing high-dimensional mapping of CEACAM1 expression, researchers should consider:

  • Antibody validation: Use antibodies that distinguish human CEACAM1 from other highly related CEACAM family members. For example, researchers have successfully used antibodies labeled with 159Tb in CyTOF panels .

  • Comprehensive panel design: Include antibodies for CEACAM1 alongside other relevant markers such as PD1 and PD-L1 (targets of immunotherapy) to create informative immune cell atlases. This approach allows for correlation of CEACAM1 expression with other clinically relevant markers .

  • Sample collection strategy: Analyze both peripheral blood and tumor samples from patients to compare CEACAM1 expression patterns. Include treatment-naïve and treatment-resistant cases, as well as healthy controls for comprehensive analysis .

  • Cell population identification: Analyze CEACAM1 expression across diverse immune cell populations, including NK cells, innate T cells, B cells, monocytic cells, dendritic cells, CD4+ T cells, and CD8+ T cells to create a complete expression atlas .

  • Data analysis approaches: Employ computational methods to identify discrete populations and quantify co-expression patterns. This has revealed, for example, that the majority of circulating PD1-positive immune cells in treatment-resistant melanoma co-express CEACAM1 .

How should researchers approach studying the various CEACAM1 isoforms in different experimental contexts?

Isoform-specific research approaches for CEACAM1 should include:

  • Isoform-specific primers and probes: Design RT-PCR primers that can distinguish between the 12 alternatively spliced isoforms, particularly differentiating between those with long (L) or short (S) cytoplasmic domains .

  • Functional domain analysis: Consider the specific functions of different domains - the IgV domain is critical for ligand interactions, while the cytoplasmic domain (particularly the ITIM motifs in L variants) is essential for signaling .

  • Secreted vs. membrane-bound forms: Account for both membrane-integrated isoforms (like CEACAM1-4L, CEACAM1-1S) and secreted proteins (CEACAM1-3, CEACAM1-3C2) that lack transmembrane sequences .

  • Isoform-specific antibodies: When possible, use antibodies that can distinguish between isoforms or at least between the major structural variants (4L, 4S, 2L, 2S, etc.) .

  • Context-dependent expression analysis: Different isoforms may be preferentially expressed in different tissues or disease states. For example, melanoma cell lines expressing higher levels of CEACAM1-3L and CEACAM1-3S splice variants show greater response to CEACAM1-targeting treatments .

What are the emerging techniques for therapeutic targeting of CEACAM1 in disease?

Emerging approaches for therapeutic targeting of CEACAM1 include:

  • Human antibody fragments: Fully human antibody fragments like DIATHIS1, developed through biopanning from phage antibody libraries, represent promising biotechnological platforms for antimelanoma therapeutics .

  • Combination therapies: Given CEACAM1's correlation with treatment resistance and its co-expression with PD1 on various immune cells, combining CEACAM1-targeting agents with established checkpoint inhibitors may overcome resistance mechanisms .

  • Isoform-specific targeting: Developing therapeutics that specifically target certain CEACAM1 isoforms (like CEACAM1-3L and CEACAM1-3S) might provide more precise approaches for melanoma treatment .

  • Host-pathogen interaction blockade: Agents that disrupt the interaction between bacterial proteins (like Streptococcus pyogenes R28) and human CEACAM1 could potentially prevent or treat serious infections such as puerperal sepsis .

  • Domain-specific targeting: Strategies focused on blocking specific functional domains (like the IgV domain responsible for homophilic interactions) rather than targeting the entire protein may offer more selective therapeutic approaches with fewer off-target effects .

Product Science Overview

Structure and Function

CEACAM1 is a glycoprotein that mediates cell adhesion via both homophilic (binding to itself) and heterophilic (binding to other proteins) interactions . It plays a crucial role in various biological processes, including:

  • Cell Adhesion: CEACAM1 facilitates cell-cell adhesion, which is essential for maintaining tissue architecture and integrity .
  • Immune Response: It is involved in the regulation of immune responses, acting as a co-inhibitory molecule that can modulate T-cell responses .
  • Angiogenesis: CEACAM1 is associated with early stages of angiogenesis, regulating the proliferation, migration, and differentiation of endothelial cells .
Clinical Significance

CEACAM1 has been identified as a prognostic marker in various cancers, including colorectal, lung, and pancreatic cancers . Its expression is often altered in tumor cells, and it can influence tumor progression and metastasis . The protein’s role in cancer has made it a target for therapeutic interventions and a biomarker for disease monitoring .

Recombinant CEACAM1

Recombinant CEACAM1 is produced using recombinant DNA technology, which involves inserting the gene encoding CEACAM1 into a suitable expression system, such as bacteria or mammalian cells. This allows for the large-scale production of the protein for research and therapeutic purposes.

Research and Applications

Research on CEACAM1 has provided insights into its diverse functions and its role in disease. Studies have shown that CEACAM1 expression can be modulated by various factors, including cytokines and bacterial infections . For example, non-typable Haemophilus influenzae and Moraxella catarrhalis can increase CEACAM1 expression in bronchial epithelial cells .

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.