CADM1 Human

Cell Adhesion Molecule 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CADM1

Cell Adhesion Molecule 1 (CADM1), also known as TSLC1, IGSF4, and SynCAM, is a transmembrane glycoprotein belonging to the immunoglobulin superfamily (IgSF). It plays pivotal roles in cell adhesion, signal transduction, and tumor suppression across multiple biological systems .

Cell Adhesion and Morphology

CADM1 mediates adhesion between adjacent cells via trans-homophilic interactions, stabilizing epithelial integrity and polarity . In mast cells, it accounts for 67% of adhesion to human lung fibroblasts and 32% to airway smooth muscle cells, promoting survival and proliferation .

Cell TypeAdhesion TargetCADM1 ContributionReference
HMC-1 (mast cells)Lung fibroblasts~67%
HLMCs (mast cells)Airway smooth muscle cells~32%
Pancreatic β cellsIslet myeloid cellsImmune cell recruitment

Signaling Pathways

  • Hippo Pathway: Recruits MST1/2 and LATS1/2 kinases to inhibit YAP1, suppressing proliferation in lung adenocarcinoma .

  • PI3K/AKT/mTOR: Activates cytoskeleton reorganization and epithelial structure formation .

  • c-Src Regulation: Binds Cbp to recruit Csk, phosphorylating c-Src Y529 and inhibiting its oncogenic activity .

Neurodevelopmental and Immune Functions

  • ADHD and Autism: Mutations in CADM1 (e.g., C739A, A755C) impair synaptogenesis, dendritic growth, and ER stress responses, linking to neurodevelopmental disorders .

  • Type 1 Diabetes: Enriched in islet myeloid cells (macrophages, DCs), facilitating immune cell infiltration and β-cell destruction .

Tumor Suppression

CADM1 acts as a tumor suppressor in multiple cancers:

Prognostic Biomarker

Cancer TypeCADM1 ExpressionOutcomeReference
cSCCHighFavorable prognosis
Ovarian/Bladder CancerOverexpressionIncreased apoptosis

Tissue Expression Profile

CADM1 is highly expressed in:

TissuePrimary ExpressionFunctional RoleReference
LungEpithelial cellsTumor suppression
Pancreasβ cells, myeloid cellsImmune regulation
BrainPrefrontal lobeSynaptic plasticity
SkincSCC tumorsPrognostic marker

Mast Cell Biology

RNA interference (shRNA) targeting CADM1 reduces mast cell adhesion to fibroblasts by ~50%, underscoring its role in inflammatory microenvironments .

Neurological Disorders

Polymorphisms (e.g., rs10891819) in CADM1 intron 9 correlate with higher prefrontal activity and enhanced inhibitory control in ADHD, suggesting a protective role .

Protein Characteristics

PropertyDetailReference
Molecular Weight37.9 kDa (calculated)
GlycosylationSDS-PAGE bands: 50–85 kDa
Binding AffinityBinds CRTAM with 0.6–5 ng/mL EC₅₀

Product Specs

Introduction
Cell Adhesion Molecule 1, also known as CADM1, is a member of the nectin family. This molecule facilitates both homophilic cell-cell adhesion in a calcium-independent manner and heterophilic cell-cell adhesion with CADM3 and PVRL3, also in a calcium-independent manner. CADM1 acts as a tumor suppressor in non-small-cell lung cancer (NSCLC) cells. Its interaction with CRTAM promotes natural killer (NK) cell cytotoxicity and interferon gamma (IFN-γ) secretion by CD8+ cells in vitro. This interaction also enhances NK cell-mediated rejection of tumors expressing CADM3 in vivo.
Description
Recombinant human CADM1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 353 amino acids (45-374 a.a.). It has a molecular mass of 39.4 kDa. The protein includes a 23 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
The CADM1 protein solution is provided at a concentration of 0.25 mg/ml and contains phosphate-buffered saline (pH 7.4), 10% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freeze-thaw cycles should be avoided.
Purity
Purity is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
Cell Adhesion Molecule 1, Tumor Suppressor In Lung Cancer 1, IGSF4, Spermatogenic Immunoglobulin Superfamily, Immunoglobulin Superfamily, Member 4, Immunoglobulin Superfamily Member 4, Synaptic Cell Adhesion Molecule, Nectin-Like Protein 2, Nectin-Like 2, Necl-2, SgIGSF, IGSF4A, SYNCAM, TSLC-1, TSLC1, NECL2, Immunoglobulin Superfamily, Member 4D Variant 1, Immunoglobulin Superfamily, Member 4D Variant 2, TSLC1/Nectin-Like 2/IGSF4, Truncated CADM1 Protein, STSLC-1, SynCAM1, RA175, ST17, BL2, Cell adhesion molecule 1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSQNLFTKD VTVIEGEVAT ISCQVNKSDD SVIQLLNPNR QTIYFRDFRP LKDSRFQLLN FSSSELKVSL TNVSISDEGR YFCQLYTDPP QESYTTITVL VPPRNLMIDI QKDTAVEGEE IEVNCTAMAS KPATTIRWFK GNTELKGKSE VEEWSDMYTV TSQLMLKVHK EDDGVPVICQ VEHPAVTGNL QTQRYLEVQY KPQVHIQMTY PLQGLTREGD ALELTCEAIG KPQPVMVTWV RVDDEMPQHA VLSGPNLFIN NLNKTDNGTY RCEASNIVGK AHSDYMLYVY DPPTTIPPPT TTTTTTTTTT TTILTIITDS RAGEEGSIRA VDH.

Q&A

What is the basic structure of CADM1 protein in humans?

CADM1 (Cell Adhesion Molecule 1) belongs to the immunoglobulin superfamily and features a complex structural organization essential to its function. The protein comprises three extracellular immunoglobulin loops, a single transmembrane domain, and a short C-terminal cytoplasmic domain. The cytoplasmic region contains two conserved protein interaction modules: the 4.1 binding motif and a type II PDZ-binding motif that interact with 4.1 proteins and membrane-associated guanylate kinase homologs (MAGuKs), respectively . These structural elements enable CADM1 to participate in cell adhesion, formation of epithelial cell morphology, polarity establishment, and intracellular signal transduction . CADM1 is strategically located on the lateral side of epithelial cell membranes where it mediates adhesion with neighboring cells through trans-homophilic interactions, forming the foundation for its diverse biological roles .

What are the primary expression patterns of CADM1 in human tissues?

CADM1 exhibits a distinctive expression pattern in the human body, with particularly high expression in neural tissues. Research has demonstrated that CADM1 is abundantly expressed in multiple brain regions, including the cingulate cortex, parietal lobe, temporal lobe, occipital lobe, amygdala, caudate nucleus, cerebellum, and most notably, the prefrontal cortex . This expression pattern overlaps significantly with brain regions implicated in neuropsychiatric conditions such as ADHD, suggesting functional relevance to cognitive and behavioral regulation . Beyond neurological tissues, CADM1 is also found in various epithelial tissues where it contributes to cell-cell adhesion. The differential expression across tissues indicates tissue-specific roles and potential functional specialization of CADM1 depending on its cellular context.

How does CADM1 contribute to cellular adhesion mechanisms?

CADM1 facilitates cellular adhesion through multiple molecular mechanisms. Primarily, CADM1 establishes trans-homophilic interactions between adjacent cells, serving as a molecular bridge that promotes intercellular adhesion . At the molecular level, CADM1 forms a multiprotein complex at the cell periphery by associating with scaffold proteins including MPP3, DIg, membrane-associated guanylate kinase homolog (MAGuK) proteins, and PI3K . This complex activates the PI3K pathway, resulting in actin cytoskeleton reorganization and proper formation of epithelial cell structure . In human mast cells, CADM1 isoforms demonstrably affect cellular adhesion to different human cell types - for example, transfection of SP1 and SP6 isoforms reduced adhesion of HMC-1 cells to human lung fibroblasts but not to airway smooth muscle cells . These mechanisms collectively contribute to tissue organization, cellular communication, and maintaining structural integrity in various physiological contexts.

What is the relationship between CADM1 and attention deficit hyperactivity disorder (ADHD)?

Research has established significant connections between CADM1 and ADHD through multiple lines of evidence. Genetic studies have identified CADM1 as a candidate gene potentially involved in ADHD pathophysiology, joining other cell adhesion-related genes like ITGA1 and CDH13 . Animal models provide compelling support for this connection - mice with dominant negative mutations in CADM1 (GFAP-DNSynCAM1) display several ADHD-like behaviors including increased daytime activity, decreased rest, nocturnal hyperactivity, and heightened impulsivity and aggression . Importantly, these behavioral anomalies were reversed by amphetamine administration, paralleling clinical responses seen in human ADHD treatment .

The neurobiological basis for this relationship likely involves CADM1's high expression in brain regions implicated in ADHD pathophysiology. Functional neuroimaging studies have identified overlap between brain regions with high CADM1 expression and those showing altered activity in ADHD, particularly regions involved in inhibition, working memory, and set-shifting . Based on the Research Domain Criteria (RDoC) approach, exploring the "gene-brain-behavior" relationships of CADM1 may provide more precise understanding of its role in ADHD than traditional diagnostic categories alone .

How can researchers effectively study CADM1 function in neuropsychiatric conditions?

Studying CADM1 in neuropsychiatric contexts requires a multifaceted approach integrating genetic, neuroimaging, cognitive, and behavioral methodologies. Researchers should consider implementing:

  • Genetic approaches: Tag single nucleotide polymorphism (SNP) analysis can identify relevant CADM1 variants in patient cohorts compared to controls. The search results describe a study utilizing 10-tag SNPs in 1,040 children/adolescents with ADHD and 963 controls for case-control association analyses .

  • Neurocognitive assessment: Specific cognitive domains should be evaluated using standardized tasks. For example, researchers can assess inhibition using the Stroop color-word interference test, working memory using the Rey-Osterrieth complex figure test, and set-shifting using the trail making test .

  • Neuroimaging methods: Functional magnetic resonance imaging (fMRI) is valuable for examining brain activity patterns. Resting-state fMRI with metrics such as mean amplitude of low-frequency fluctuations (mALFF) can identify alterations in brain regions with high CADM1 expression .

  • RDoC framework application: Following the Research Domain Criteria approach allows examination of CADM1 across multiple domains from genes to behavior, providing more granular insights than traditional diagnostic categories alone .

  • Integration of findings: Correlating genetic variations with brain activity patterns and cognitive/behavioral measures can establish mechanistic relationships between CADM1 function and neuropsychiatric phenotypes.

This methodological integration can yield findings with greater specificity than conventional phenotype-based approaches alone.

What is the significance of CADM1 in glioblastoma and other central nervous system tumors?

CADM1 plays a critical role in central nervous system tumors, particularly glioblastoma (GBM), where it demonstrates significant clinical and prognostic relevance. Research has established that CADM1 expression is markedly reduced in glioblastoma tissues compared to normal brain tissues . Both CADM1 protein and mRNA levels are downregulated in GBM patient tumor samples, with evidence suggesting that this suppression is regulated by exosomal miR-148a .

Clinically, CADM1 expression patterns correlate with patient outcomes. Lower CADM1 expression is associated with poor median survival and cancer progression in GBM patients, indicating CADM1's potential utility as a prognostic biomarker . This relationship suggests CADM1 may have essential functions in regulating glioblastoma proliferation and metastasis.

In neuroblastoma, another common nervous system tumor primarily affecting children, CADM1 similarly demonstrates prognostic significance. Higher CADM1 expression correlates significantly with improved survival rates . Loss of CADM1 in chromosome region 11q23 is linked to poor prognosis in neuroblastoma patients . Clinicopathological analyses have confirmed that CADM1 expression levels significantly correlate with neuroblastoma stage and pathological classification according to Shimada criteria .

These findings collectively establish CADM1 as an important molecular marker in nervous system tumors and suggest its potential as a therapeutic target requiring further investigation.

What are the major functional isoforms of CADM1 identified in humans?

Research has identified multiple functional isoforms of CADM1 in human tissues, particularly in mast cells. Four primary functional isoforms have been characterized: SP4, SP1, SP6, and SP3, which result from alternative splicing events between exons 7 and 11 . These functional isoforms play distinct roles in cellular processes, particularly in cell adhesion and signaling.

In human mast cells, these isoforms demonstrate differential expression and functional properties. Notably, the longer isoforms SP1 and SP6 are predominantly expressed in differentiated human lung mast cells (HLMCs) . Functional studies have revealed that these isoforms exhibit distinct adhesion properties - transfection experiments showed that SP1 and SP6, but not SP4, reduced adhesion of HMC-1 cells (a human mast cell line) to human lung fibroblasts, though interestingly, they did not affect adhesion to airway smooth muscle cells . This differential effect suggests isoform-specific roles in mediating cell-cell interactions in different tissue contexts.

The functional diversity of these isoforms is further reflected in their protein expression patterns. Highly glycosylated CADM1 (approximately 105 kDa) has been detected by western blotting in mast cells, while an extracellular domain (approximately 95 kDa) was found only in the culture medium from HLMCs but not HMC-1 cells, indicating differential protein expression and processing .

How do dysfunctional CADM1 isoforms arise and what are their implications?

Dysfunctional CADM1 isoforms arise primarily through alternative splicing mechanisms involving cryptic exons. Research has identified two dysfunctional isoforms in human mast cells that result from alternative splicing of cryptic exons A and B located between exons 1 and 2 of the CADM1 gene . These cryptic exon inclusions introduce premature termination codons that truncate the protein, leading to non-functional CADM1 variants.

The prevalence of these dysfunctional isoforms is significant - they have been detected in approximately 40% of mast cell specimens examined . Intriguingly, genomic DNA sequencing analyses have revealed that the splicing of cryptic exon B does not result from specific single nucleotide polymorphisms (SNPs) within this exon or its putative splice branch point , suggesting other regulatory mechanisms control this alternative splicing event.

The implications of these dysfunctional isoforms are considerable for CADM1 biology and function. Their presence may act as a natural dominant-negative mechanism that modulates CADM1 activity in cells. This could represent an additional layer of regulation for CADM1 function, potentially influencing cell adhesion, signaling, and other CADM1-dependent processes. In pathological contexts, the balance between functional and dysfunctional isoforms may be disrupted, contributing to disease states where CADM1 function is compromised.

What experimental approaches are optimal for studying CADM1 isoform expression and function?

To comprehensively study CADM1 isoform expression and function, researchers should employ a multifaceted methodological approach:

  • Isoform Identification and Quantification:

    • RT-PCR with isoform-specific primers targeting the differentially spliced regions between exons 7-11 can identify functional isoforms (SP4, SP1, SP6, SP3) .

    • Nested PCR approaches may enhance detection sensitivity for low-abundance isoforms.

    • Real-time quantitative PCR allows precise quantification of relative isoform abundance.

    • RNA sequencing provides comprehensive detection of all splice variants, including potentially novel isoforms.

  • Protein Expression Analysis:

    • Western blotting with glycosylation-sensitive detection methods can distinguish the approximately 105 kDa glycosylated CADM1 from other forms .

    • Immunoprecipitation followed by mass spectrometry enables detailed characterization of post-translational modifications specific to each isoform.

    • Analysis of culture medium for secreted/shed extracellular domains (~95 kDa) provides insights into differential processing between cell types .

  • Functional Characterization:

    • Transfection studies with individual isoforms in relevant cell lines (e.g., HMC-1) followed by adhesion assays to different cell types (e.g., fibroblasts, smooth muscle cells) reveal isoform-specific functional differences .

    • Co-immunoprecipitation experiments identify isoform-specific protein interaction partners.

    • CRISPR/Cas9-mediated isoform-specific knockout or mutation studies assess physiological roles.

    • Conditional expression systems enable temporal control of isoform expression for studying developmental aspects.

  • Clinical Correlation:

    • Analysis of isoform expression patterns in patient samples can establish clinicopathological correlations.

    • Single-cell RNA sequencing of affected tissues can reveal cell type-specific isoform expression patterns in disease states.

Integration of these approaches provides comprehensive insights into the complex roles of CADM1 isoforms in normal physiology and disease.

What molecular mechanisms underlie CADM1's tumor suppressor function?

CADM1 exerts its tumor suppressor functions through multiple molecular mechanisms that regulate cell proliferation, adhesion, and apoptosis. These mechanisms include:

  • Regulation of the Hippo pathway: CADM1 participates in the Hippo signaling pathway, which controls cell proliferation and contact inhibition. It forms a scaffold protein complex with NF2, KIBRA, and SAV1, which recruits MST1/2 and LATS1/2 kinases to the cell membrane. This activation of the Hippo pathway leads to phosphorylation events that inactivate YAP1 and inhibit the transcription of its target genes, ultimately suppressing cell proliferation .

  • Cytoplasmic domain-mediated tumor suppression: CADM1 relies on its cytoplasmic domain for its anti-tumorigenic effects. Through its 4.1 binding motif and interaction with 4.1B/DAL1, CADM1 can inhibit tumorigenicity in experimental models. Additionally, it participates in cbp-dependent c-Src regulation, which contributes to its tumor suppressor function .

  • c-Src pathway modulation: CADM1 forms a CADM1-cbp-c-Src complex that regulates c-Src activity. Cbp (Csk binding protein) recruits Csk to lipid rafts, where Csk phosphorylates c-Src at Y529, inactivating its catalytic activity. This mechanism is particularly relevant in colon cancer, where c-Src is highly activated and contributes to malignant transformation .

  • Cell death induction under non-adhesive conditions: In malignant melanoma, elevated CADM1 can induce cell death through multiple pathways. It increases PARP-1 (poly ADP ribose polymerase) and recruits AIF (mitochondrial apoptosis inducing factor) and MIF (microphage migration inhibitory factor) to the nucleus, breaking down DNA into large fragments. Additionally, it can induce the destruction of mitochondrial membrane potential, leading to cell-dependent cell death .

  • Inhibition of epithelial-mesenchymal transition (EMT): CADM1 can inhibit the EMT process by suppressing TWIST1 activation through inhibition of the RAS-ERK pathway. This mechanism prevents cancer cell invasion and metastasis .

These diverse mechanisms collectively contribute to CADM1's role as a tumor suppressor across multiple cancer types.

How does CADM1 expression correlate with clinical outcomes in different cancer types?

CADM1 expression demonstrates significant correlations with clinical outcomes across multiple cancer types, with substantial implications for prognosis and treatment strategies:

  • Glioblastoma (GBM): Low CADM1 expression in GBM correlates with poor median survival and accelerated cancer progression, establishing CADM1 as a negative prognostic indicator . CADM1 downregulation appears to be regulated by exosomal miR-148a, suggesting potential therapeutic strategies targeting this regulatory mechanism .

  • Neuroblastoma: High CADM1 expression is significantly associated with improved survival rates in neuroblastoma patients . Loss of CADM1 in chromosome region 11q23 correlates with poor prognosis, and immunohistochemical analysis confirms that low CADM1 expression predicts unfavorable outcomes . CADM1 expression levels also correlate significantly with neuroblastoma stage and Shimada classification, reinforcing its clinical relevance .

  • Cervical cancer: CADM1 serves as a biomarker for assessing cervical epithelial lesions, with lower CADM1 levels typically associated with more severe cervical lesions . This correlation makes CADM1 a valuable marker for disease progression in cervical neoplasia.

  • Adult T-cell leukemia/lymphoma (ATLL): Unlike most cancers, CADM1 is overexpressed in ATLL cells and serves as a surface marker for human T-cell leukemia virus (HTLV-1) infection of T cells . Clinical data from 71 cases demonstrates that HTLV-1-infected individuals can be stratified into four prognostic groups based on the percentage of CD4+ CADM1+ cells: G1 (CADM1+ ≤10%), G2 (10% < CADM1+ ≤25%), G3 (25% < CADM1+ ≤50%), and G4 (50% < CADM1+), with G4 patients exhibiting the highest risk - 28.4% requiring systemic chemotherapy within 3 years .

These correlations establish CADM1 as a clinically relevant biomarker across multiple malignancies, with potential applications in diagnosis, prognosis, and treatment selection.

What methodological approaches should be used to investigate CADM1's role in tumor progression and metastasis?

A comprehensive investigation of CADM1's role in tumor progression and metastasis requires integration of multiple methodological approaches:

  • Expression Analysis in Clinical Samples:

    • Immunohistochemistry to assess CADM1 protein expression levels in tumor samples versus adjacent normal tissues

    • RT-qPCR for CADM1 mRNA quantification across tumor stages and grades

    • Methylation-specific PCR to evaluate CADM1 promoter methylation status, as epigenetic silencing is a common mechanism of CADM1 downregulation in malignancies

    • Western blotting with glycosylation analysis to detect potential alterations in post-translational modifications

  • Functional Studies in Cell Models:

    • CRISPR/Cas9-mediated CADM1 knockout or overexpression in cancer cell lines

    • Domain-specific mutations to isolate functions of cytoplasmic versus extracellular regions

    • Cell adhesion assays to quantify effects on homotypic and heterotypic cell interactions

    • Migration and invasion assays (transwell, scratch/wound healing) to assess metastatic potential

    • Apoptosis assays under adherent and non-adherent conditions to evaluate anoikis resistance

    • Co-culture systems with endothelial cells to study tumor-endothelium interactions

  • Pathway Analysis:

    • Co-immunoprecipitation to identify CADM1 interaction partners in tumor versus normal cells

    • Phosphorylation status analysis of downstream effectors in the Hippo pathway (YAP/TAZ)

    • Assessment of c-Src activation status through phospho-specific antibodies targeting Y418 and Y529

    • EMT marker analysis (E-cadherin, vimentin, TWIST1) following CADM1 modulation

  • In Vivo Models:

    • Xenograft models with CADM1-modulated cancer cells to assess tumor growth kinetics

    • Metastasis models to evaluate organ-specific spread

    • Treatment studies with anti-CADM1 antibodies or epigenetic modifiers like OD-2100

    • Patient-derived xenografts to maintain tumor heterogeneity

  • Molecular Interaction Studies:

    • Analysis of CADM1 interactions with specific miRNAs (miR-1246, miR-205) implicated in its regulation

    • Investigation of interactions with other surface receptors like HER2, which can form complexes with CADM1

Integration of these approaches provides comprehensive insights into CADM1's context-dependent roles in tumor biology, potentially identifying novel therapeutic strategies targeting CADM1-dependent mechanisms.

How does CADM1 interact with major intracellular signaling cascades?

CADM1 interacts with multiple intracellular signaling cascades through its cytoplasmic domain, serving as a critical node that integrates cell adhesion with downstream signaling:

  • PI3K-Akt Pathway: CADM1 forms a multiprotein complex with MPP3, DIg, membrane-associated guanylate kinase homolog (MAGuK) proteins, and PI3K at the cell periphery . This complex activation triggers the PI3K pathway, leading to actin cytoskeleton reorganization and proper epithelial cell structure formation . In cancer contexts, modulation of this pathway affects cell cycle progression, with CADM1-mediated upregulation of PTEN inhibiting AKT and GSK-3β phosphorylation .

  • Hippo Signaling Pathway: CADM1 serves as a scaffold for the Hippo pathway components, forming complexes with NF2, KIBRA, and SAV1 . This complex recruits MST1/2 and LATS1/2 kinases to the cell membrane, activating the Hippo pathway. The subsequent phosphorylation events inactivate YAP1, inhibiting the transcription of its target genes and suppressing cell proliferation .

  • RAS-ERK Pathway: In melanoma cells, CADM1 inhibits the RAS-RAF-MEK1/2-ERK1/2 signaling cascade, which prevents activation of TWIST1, a transcription factor promoting epithelial-mesenchymal transition (EMT) . Through this mechanism, CADM1 suppresses cancer cell invasion and migration.

  • c-Src Regulation: CADM1 participates in cbp-dependent c-Src regulation through the formation of a CADM1-cbp-c-Src complex . Cbp recruits Csk to lipid rafts, where Csk phosphorylates c-Src at Y529, inactivating its catalytic activity and inhibiting downstream oncogenic signaling .

  • STAT3 Signaling: CADM1 can interact with HER2 through its extracellular domain, activating the downstream STAT3 signaling pathway . This CADM1/HER2/STAT3 axis has been implicated in tumor metastasis, suggesting context-dependent roles for CADM1 in cancer progression .

  • Cell Cycle Regulation: CADM1-mediated signaling affects the expression of cell cycle-related proteins, including cyclin D, cyclin E, CDK2, CDK4, and CDK6 (downregulated) and p15, p21, and p27 (upregulated) . These changes lead to G0/G1 phase arrest and inhibition of cell proliferation through the formation of p15-cyclinD/CDK4/CDK6 or p21/p27-cyclinE/CDK2 complexes .

These interactions highlight CADM1's role as a multifunctional signaling mediator that can influence diverse cellular processes through its connections to major signaling networks.

What is the relationship between CADM1 and the NF-κB signaling pathway in Adult T-cell Leukemia/Lymphoma (ATLL)?

The relationship between CADM1 and the NF-κB signaling pathway in Adult T-cell Leukemia/Lymphoma (ATLL) represents a critical paradigm in understanding this aggressive malignancy:

CADM1 demonstrates a unique expression pattern in ATLL compared to other malignancies. While CADM1 typically functions as a tumor suppressor with reduced expression in most cancers, it is characteristically overexpressed in ATLL cells . This overexpression serves as a distinctive surface marker for human T-cell leukemia virus (HTLV-1) infection of T cells, the causative agent of ATLL .

The NF-κB signaling pathway is believed to be integrally involved in ATLL pathogenesis . HTLV-1 infection activates NF-κB signaling through multiple viral factors, particularly the viral protein Tax. The sustained activation of NF-κB promotes cell survival, proliferation, and resistance to apoptosis in ATLL cells.

Functional studies have demonstrated that overexpressed CADM1 in ATLL cells promotes several oncogenic processes:

  • Enhanced self-aggregation of ATLL cells

  • Increased adhesion to endothelial cells

  • Augmented tumor growth and invasion in xenograft mice models

The clinical relevance of this relationship is substantial. Analysis of 71 clinical cases revealed that HTLV-1-infected individuals can be stratified into four prognostic groups based on the percentage of CD4+ CADM1+ cells:

  • G1 (CADM1+ ≤10%): Stable condition

  • G2 (10% < CADM1+ ≤25%): Stable condition

  • G3 (25% < CADM1+ ≤50%): Increased risk

  • G4 (50% < CADM1+): Highest risk, with 28.4% requiring systemic chemotherapy within 3 years

These findings suggest potential therapeutic avenues. The DNA hypermethylation inhibitor OD-2100 has shown efficacy in anti-ATLL activity and inhibition of tumor growth . Additionally, anti-CADM1 antibodies have demonstrated the ability to inhibit interactions between endothelial cells and CADM1+ ATLL cells, potentially suppressing tumor metastasis .

This unique relationship between CADM1 and NF-κB signaling in ATLL highlights the context-dependent roles of CADM1 and suggests targeted therapeutic approaches for this aggressive malignancy.

How do miRNAs regulate CADM1 expression and function in different biological contexts?

MicroRNAs (miRNAs) play crucial roles in post-transcriptional regulation of CADM1 expression across various biological contexts, creating a complex regulatory network with significant implications for normal physiology and disease:

  • Direct miRNA targeting mechanisms:

    • miR-1246 directly targets CADM1 by binding to its 3'UTR, suppressing CADM1 expression and promoting invasive migration of hepatocellular carcinoma (HCC) cells in vitro .

    • miR-205 similarly targets CADM1 in cervical cancer, with studies demonstrating that downregulation of miR-205 inhibits cervical cancer cell invasion and angiogenesis through the CADM1-mediated Akt signaling pathway. This mechanism has been validated in both in vitro experiments and in vivo nude mouse models .

  • Context-specific miRNA regulation:

    • In glioblastoma, exosomal miR-148a regulates CADM1 expression levels. Low expression of CADM1 protein and mRNA in GBM tumor tissues is mediated by this miRNA regulatory mechanism .

    • Multiple miRNAs have been identified that target CADM1 across different cancer types, creating a comprehensive regulatory network that influences tumor proliferation and invasion of multiple systems .

  • Integrated regulatory networks:

    • The relationship between CADM1 and related miRNAs operates through corresponding pathways to regulate tumor biology. For example, miRNA-mediated suppression of CADM1 can affect downstream signaling through the Hippo, PI3K-AKT, and RAS-ERK pathways .

    • These regulatory relationships are not static but dynamic, responding to cellular context and environmental signals.

  • Therapeutic implications:

    • Understanding the miRNA regulation of CADM1 provides potential therapeutic opportunities. Strategies targeting specific miRNAs (anti-miRs) could potentially restore CADM1 expression in cancers where it functions as a tumor suppressor.

    • Conversely, in contexts like ATLL where CADM1 is overexpressed, miRNA mimics targeting CADM1 might offer therapeutic benefits.

The complex interplay between miRNAs and CADM1 highlights the intricate regulatory mechanisms controlling CADM1 expression and function. These mechanisms provide insights into the contextual role of CADM1 in different biological systems and offer potential targets for therapeutic intervention in CADM1-associated diseases.

What are the optimal techniques for detecting CADM1 expression in different human tissues?

Detecting CADM1 expression across different human tissues requires a strategic combination of techniques that address the protein's unique characteristics, including its multiple isoforms, post-translational modifications, and tissue-specific expression patterns:

  • Transcriptomic Detection Methods:

    • RT-PCR with isoform-specific primers: Design primers targeting junctions between exons 7-11 to distinguish between functional isoforms SP4, SP1, SP6, and SP3 . Additional primers targeting the region between exons 1-2 can detect dysfunctional isoforms containing cryptic exons A and B .

    • Quantitative Real-Time PCR (qRT-PCR): For precise quantification of CADM1 mRNA levels across different tissues, with normalization to appropriate housekeeping genes for the specific tissue type being studied.

    • RNA-Seq: Provides comprehensive detection of all CADM1 transcript variants, including potentially novel isoforms, with digital quantification capabilities.

    • Single-cell RNA sequencing: Offers insights into cell-type specific expression patterns within heterogeneous tissues, particularly valuable for brain tissues where cellular heterogeneity is pronounced .

  • Protein Detection Methods:

    • Western blotting: Requires careful consideration of CADM1's extensive glycosylation - highly glycosylated CADM1 appears at approximately 105 kDa, while the extracellular domain fragment at approximately 95 kDa may be detected in culture supernatants . Use of deglycosylation enzymes may be necessary to resolve ambiguous bands.

    • Immunohistochemistry (IHC): Critical for visualizing spatial distribution of CADM1 within tissue architecture. Selection of antibodies recognizing conserved epitopes across isoforms is essential for comprehensive detection.

    • Immunofluorescence: Offers superior resolution for subcellular localization, particularly important for examining CADM1's distribution at lateral cell membranes in polarized epithelial cells .

    • Flow cytometry: Valuable for quantifying CADM1 expression in specific cell populations, particularly in hematological tissues. This technique has proven particularly useful in ATLL research, where CD4+CADM1+ percentages provide clinical stratification .

  • Specialized Applications:

    • Methylation-specific PCR: Essential for detecting epigenetic silencing of CADM1, a common mechanism in cancer tissues .

    • Chromatin immunoprecipitation (ChIP): Identifies transcription factors regulating CADM1 expression in different tissue contexts.

    • Proximity ligation assay: Detects CADM1 interactions with binding partners in situ, providing insights into tissue-specific protein complexes.

The optimal approach typically combines multiple techniques to provide complementary data on CADM1 expression, localization, and functional status across different human tissues.

What are the challenges in studying CADM1 protein-protein interactions and how can they be overcome?

Studying CADM1 protein-protein interactions presents several unique challenges requiring specialized methodological approaches:

  • Transmembrane Protein Complexities:

    • Challenge: CADM1's transmembrane nature makes it difficult to preserve native interactions during isolation.

    • Solution: Use membrane-compatible detergents (e.g., digitonin, CHAPS) for gentle solubilization while maintaining protein-protein interactions. Crosslinking approaches with membrane-permeable crosslinkers (DSP, formaldehyde) can stabilize transient interactions before extraction.

  • Glycosylation Interference:

    • Challenge: CADM1's extensive glycosylation (appearing at ~105 kDa) can interfere with interaction studies by masking binding sites or creating steric hindrance .

    • Solution: Strategic deglycosylation using endoglycosidases (PNGase F, Endo H) can be employed to remove N-glycans while preserving protein structure. Alternatively, expression of CADM1 in glycosylation-deficient cell lines can provide cleaner interaction data.

  • Isoform-Specific Interactions:

    • Challenge: Different CADM1 isoforms (SP4, SP1, SP6, SP3) likely have distinct interaction profiles that must be distinguished .

    • Solution: Express individual tagged isoforms in cellular models followed by affinity purification. Mass spectrometry-based comparative interactomics can identify isoform-specific binding partners. Domain-specific antibodies can be used to immunoprecipitate specific variants.

  • Homophilic vs. Heterophilic Interactions:

    • Challenge: CADM1 forms both homophilic (CADM1-CADM1) and heterophilic interactions, making it difficult to distinguish interaction types.

    • Solution: Bimolecular Fluorescence Complementation (BiFC) assays with differentially tagged CADM1 molecules can visualize homophilic interactions. FRET/FLIM approaches provide spatial resolution of interaction dynamics. Single-molecule imaging techniques can resolve stoichiometry of interaction complexes.

  • PDZ and 4.1 Binding Motif Interactions:

    • Challenge: The cytoplasmic domain of CADM1 contains both PDZ-binding and 4.1-binding motifs that mediate distinct interactions with scaffolding proteins .

    • Solution: Mutational analysis targeting specific motifs allows mapping of domain-specific interactions. Peptide arrays with cytoplasmic domain fragments can identify specific binding regions. Proximity-dependent biotinylation (BioID, TurboID) can capture more transient interactions in living cells.

  • Physiological Relevance Assessment:

    • Challenge: In vitro identified interactions may not reflect physiological relevance in vivo.

    • Solution: Validate interactions in relevant primary cells rather than just cell lines. Develop knockin mouse models with tagged endogenous CADM1 for physiological interactome studies. Use tissue-specific proximity labeling approaches to capture context-dependent interactions.

  • Signaling Complex Dynamics:

    • Challenge: CADM1 participates in multiple signaling pathways (Hippo, PI3K, RAS-ERK) with dynamic, context-dependent interaction networks.

    • Solution: Temporal analysis following stimulus application can capture dynamic interaction changes. Phosphoproteomic analysis of CADM1 complexes reveals regulation by post-translational modifications. Spatial proteomics approaches can determine subcellular locations of specific interaction events.

Integration of these approaches creates a comprehensive strategy for elucidating CADM1's complex interactome across different physiological and pathological contexts.

What approaches should be used to study CADM1's functional role in cell adhesion and signal transduction?

Studying CADM1's dual roles in cell adhesion and signal transduction requires an integrated experimental approach that bridges structural, cellular, and molecular dimensions:

  • Cell Adhesion Functional Analyses:

    • Aggregation assays: Quantify CADM1-mediated homophilic adhesion by measuring cell aggregation in suspension cultures expressing different CADM1 isoforms .

    • Adhesion strength measurements: Apply atomic force microscopy (AFM) or flow-chamber detachment assays to measure the biophysical strength of CADM1-mediated adhesion.

    • Isoform-specific transfection studies: Perform comparative studies with SP1, SP4, SP6, and SP3 isoforms to determine their differential effects on adhesion to various cell types, as demonstrated in the research with HMC-1 cells adhering to lung fibroblasts versus airway smooth muscle cells .

    • 3D cell culture models: Evaluate CADM1's role in tissue architecture formation using organoid or spheroid cultures that better recapitulate in vivo cellular organization.

    • Live-cell imaging: Apply fluorescently-tagged CADM1 variants to visualize real-time dynamics of adhesion formation and turnover.

  • Signal Transduction Analysis:

    • Pathway-specific reporter assays: Implement luciferase-based reporters for PI3K-Akt, Hippo, NF-κB, and RAS-ERK pathways to quantify CADM1-mediated signaling activation .

    • Phosphoproteomic profiling: Conduct mass spectrometry-based global phosphoproteomics after CADM1 manipulation to identify all affected signaling nodes.

    • Proximity-dependent biotinylation: Apply BioID or TurboID fused to CADM1 to identify proximal proteins in the signaling nexus.

    • FRET-based biosensors: Utilize fluorescence resonance energy transfer sensors to monitor real-time activation of signaling pathways in live cells following CADM1 engagement.

    • Subcellular fractionation: Analyze redistribution of signaling components between membrane, cytoplasm, and nucleus following CADM1 activation.

  • Integrative Approaches:

    • Domain-specific mutational analysis: Create systematic mutations in extracellular, transmembrane, and cytoplasmic domains to dissect regions responsible for adhesion versus signaling functions.

    • Temporal dissection: Apply time-course analyses to determine whether adhesion precedes signaling or if these functions can be uncoupled.

    • Context-dependent studies: Compare CADM1 function across different cell types where it exhibits contrasting roles (e.g., tumor suppression in epithelial cells versus oncogenic activity in ATLL) .

    • Mechanical force studies: Investigate how mechanical forces applied to CADM1 adhesions influence downstream signaling through mechanotransduction.

    • Mathematical modeling: Develop computational models to integrate adhesion and signaling data, predicting system-level responses to CADM1 perturbations.

  • Translational Applications:

    • Patient-derived systems: Validate findings in primary cells or organoids from relevant patient populations.

    • Targeted therapeutic development: Design peptides or small molecules that selectively modulate adhesion versus signaling functions.

    • Biomarker development: Establish assays measuring CADM1-dependent adhesion or signaling as potential biomarkers for diseases like ATLL or neuropsychiatric conditions .

This comprehensive approach reveals how CADM1 integrates physical adhesion with biochemical signaling to influence cellular behavior in both physiological and pathological contexts.

How might CADM1-targeted therapies be developed for neurological and oncological conditions?

Developing CADM1-targeted therapies requires distinct approaches for neurological versus oncological conditions, reflecting CADM1's context-dependent roles as either a target for restoration (in most cancers and some neurological conditions) or inhibition (in ATLL):

A. Therapeutic Approaches for Neurological Conditions:

  • ADHD and related neurodevelopmental disorders:

    • Gene therapy approaches: Delivery of functional CADM1 to specific brain regions with high expression relevance to ADHD (prefrontal cortex, cingulate cortex) .

    • Small molecule enhancers: Development of compounds that enhance CADM1 function or expression to normalize neural circuit activity.

    • Targeted drug delivery: Nanoparticle-based approaches to deliver CADM1-modulating agents across the blood-brain barrier to affected brain regions.

    • Isoform-specific modulators: Design of therapeutics targeting specific CADM1 isoforms relevant to neurological function, potentially focusing on SP4, SP1, SP6, and SP3 variants .

  • Glioblastoma:

    • Epigenetic modifiers: Development of compounds that reverse CADM1 promoter methylation to restore expression in tumors where it's suppressed .

    • miRNA inhibitors: Design of antagomirs targeting miR-148a to counter its CADM1-suppressive effects in glioblastoma .

    • CADM1 restoration strategies: Viral-mediated delivery of CADM1 to tumor cells to reinstate its tumor suppressor functions.

B. Therapeutic Approaches for Oncological Conditions:

  • Epithelial cancers (where CADM1 functions as tumor suppressor):

    • DNA methylation inhibitors: Compounds like OD-2100 that reverse epigenetic silencing of CADM1 .

    • Pathway-targeted approaches: Pharmacological activation of downstream CADM1 pathways (Hippo, PI3K) to compensate for CADM1 loss .

    • miRNA-based therapeutics: Inhibitors of CADM1-targeting miRNAs (miR-1246, miR-205) using antisense oligonucleotides or locked nucleic acids .

    • Synthetic lethality: Identification of genes whose inhibition is selectively lethal in CADM1-deficient tumors.

  • Adult T-cell Leukemia/Lymphoma (where CADM1 is overexpressed):

    • Anti-CADM1 antibodies: Development of monoclonal antibodies that block CADM1-mediated adhesion of ATLL cells to endothelial cells, potentially reducing metastasis .

    • CADM1-targeted immunotherapies: Creation of chimeric antigen receptor (CAR) T-cells targeting CADM1 or bispecific antibodies linking CADM1+ cells to immune effectors.

    • CADM1-toxin conjugates: Antibody-drug conjugates specifically targeting CADM1-overexpressing leukemia cells.

    • Proteolysis-targeting chimeras (PROTACs): Design of bifunctional molecules that target CADM1 for proteasomal degradation.

C. Technical and Translational Considerations:

  • Delivery challenges:

    • Blood-brain barrier penetration for neurological indications requires specialized delivery systems (nanoparticles, cell-penetrating peptides).

    • Targeted delivery to specific cell populations minimizes off-target effects on normal CADM1-expressing tissues.

  • Diagnostic stratification:

    • Development of companion diagnostics to identify patients most likely to benefit from CADM1-targeted therapies.

    • Implementation of CADM1 expression profiling or CD4+CADM1+ quantification for ATLL patient stratification .

  • Combination strategies:

    • Integration with standard-of-care treatments to enhance efficacy (e.g., combining CADM1 restoration with temozolomide in glioblastoma).

    • Sequential therapeutic approaches targeting multiple aspects of CADM1 biology or downstream pathways.

These diverse therapeutic strategies highlight the potential for CADM1-targeted interventions across multiple disease contexts, with approach selection guided by CADM1's context-specific roles.

What is the potential of CADM1 as a biomarker in clinical applications?

CADM1 demonstrates significant potential as a biomarker across multiple clinical applications, with utility in diagnosis, prognosis, and treatment selection:

  • Oncology Applications:

    • Adult T-cell Leukemia/Lymphoma (ATLL): CADM1 serves as a definitive surface marker for HTLV-1 infection in T cells . Clinical research has established a robust stratification system based on the percentage of CD4+CADM1+ cells, dividing patients into four prognostic groups (G1-G4), with G4 patients (>50% CADM1+ cells) showing significantly higher risk, including 28.4% requiring systemic chemotherapy within 3 years . This stratification enables risk-adapted treatment approaches and surveillance protocols.

    • Cervical Cancer: CADM1 expression analysis facilitates assessment of cervical epithelial lesions, with lower CADM1 levels correlating with more severe cervical lesions . This association positions CADM1 as a valuable biomarker for disease progression in cervical neoplasia, potentially complementing traditional cytological screening.

    • Differential Diagnosis: CADM1 has demonstrated utility in the differential diagnosis between histologically similar tumors - for example, between osteosarcoma and chondrosarcoma . This application addresses a critical clinical need for molecular markers that can resolve diagnostic ambiguities in challenging cases.

    • Neuroblastoma: CADM1 expression levels significantly correlate with neuroblastoma stage and Shimada classification . High CADM1 expression is associated with better survival outcomes, while loss of CADM1 in chromosome region 11q23 predicts poor prognosis . This prognostic value could inform treatment intensity decisions in pediatric neuroblastoma patients.

  • Neurodevelopmental Applications:

    • ADHD and Related Conditions: Given CADM1's relationship with ADHD-like behaviors in animal models and its genetic associations in human studies , CADM1 expression patterns or genetic variants could potentially serve as biomarkers for ADHD risk, subtype classification, or treatment response prediction.

    • Brain Imaging Correlations: The relationship between CADM1 genetic variants and brain activity patterns in regions implicated in ADHD suggests potential for CADM1 genotyping to predict neuroimaging phenotypes, possibly informing personalized therapeutic approaches.

  • Technical Implementation Considerations:

    • Flow Cytometry: Quantification of CD4+CADM1+ cell percentages provides a standardized, clinically applicable approach for ATLL risk stratification .

    • Immunohistochemistry: CADM1 protein detection in tissue sections offers integration with standard pathology workflows for solid tumors.

    • Methylation Analysis: Assessment of CADM1 promoter methylation status could serve as a biomarker for epigenetic dysregulation in multiple cancer types.

    • Genetic Variant Testing: Genotyping of CADM1 SNPs associated with neuropsychiatric conditions could provide risk stratification in vulnerable populations.

    • Liquid Biopsy Potential: Detection of soluble CADM1 extracellular domain (~95 kDa) in serum could offer minimally invasive monitoring capabilities for conditions with altered CADM1 expression.

The translational implementation of CADM1 as a biomarker requires further validation in large, diverse patient cohorts, standardization of detection methods, and integration with existing clinical workflows - but the existing evidence strongly supports its potential utility across multiple clinical domains.

How can researchers design studies to investigate CADM1's role in treatment resistance mechanisms?

Designing comprehensive studies to investigate CADM1's role in treatment resistance requires multifaceted approaches that span molecular, cellular, and clinical dimensions:

  • Clinical Correlation Studies:

    • Longitudinal Biospecimen Collection: Design protocols for serial sampling of tumors before treatment, during therapy, and at progression/recurrence to track dynamic changes in CADM1 expression, localization, and isoform distribution.

    • Multi-omics Profiling: Implement integrated genomic, transcriptomic, proteomic, and epigenomic analyses to correlate CADM1 status with treatment outcomes across diverse patient cohorts.

    • Response Stratification: Categorize patient outcomes based on CADM1 expression parameters, with particular attention to differential expression of functional isoforms (SP4, SP1, SP6, SP3) and potential emergence of dysfunctional variants during treatment.

    • Statistical Methodology: Employ multivariate analysis to distinguish CADM1-specific effects from confounding factors, with Cox proportional hazards models for survival analyses and machine learning approaches for predictive biomarker development.

  • Mechanistic Laboratory Investigations:

    • Drug Resistance Models: Develop isogenic cell line pairs with differential CADM1 expression or specific isoform profiles, then subject them to incremental drug exposure to generate resistant sublines.

    • CRISPR-Based Functional Genomics: Implement genome-wide CRISPR screens in CADM1-manipulated backgrounds to identify synthetic lethal interactions that modulate treatment sensitivity.

    • Pathway Interaction Analysis: Investigate how CADM1-mediated signaling (via Hippo, PI3K-Akt, RAS-ERK pathways) intersects with known resistance mechanisms such as apoptosis evasion, DNA damage repair, or drug efflux.

    • Post-Translational Modification Studies: Examine how phosphorylation, glycosylation, or proteolytic processing of CADM1 changes in response to treatment and potentially contributes to resistance phenotypes.

    • Microenvironment Interactions: Assess how CADM1-mediated cell adhesion to stromal components might create protective niches that shield tumor cells from therapeutic agents.

  • Innovative Model Systems:

    • Patient-Derived Organoids (PDOs): Establish organoid cultures from treatment-naïve and treatment-resistant patient samples to evaluate CADM1's dynamic role in 3D models that better recapitulate tumor heterogeneity.

    • Co-Culture Systems: Develop complex co-culture models including tumor cells, immune components, and stromal elements to assess how CADM1-mediated intercellular interactions influence treatment efficacy.

    • In Vivo Resistance Models: Generate xenograft models with inducible CADM1 manipulation to assess how altering CADM1 expression or isoform distribution during treatment affects therapeutic outcomes.

    • Transgenic Mouse Models: Create conditional CADM1 knockout or isoform-specific expression models that can be crossed with disease-specific strains to evaluate treatment responses in immunocompetent settings.

  • Translational Intervention Strategies:

    • Combination Therapy Evaluation: Test whether CADM1-targeted interventions (antibodies, expression modulators) can resensitize resistant cells to standard therapies.

    • Isoform-Specific Targeting: Develop approaches to selectively modulate specific CADM1 isoforms implicated in resistance mechanisms.

    • Adaptive Trial Design: Propose biomarker-guided clinical trials that stratify patients based on CADM1 parameters and adapt treatment algorithms accordingly.

    • Pharmacodynamic Markers: Establish CADM1-related biomarkers that can be monitored during treatment to provide early indication of developing resistance.

  • Technology Implementation:

    • Single-Cell Analysis: Apply single-cell sequencing and proteomics to resolve heterogeneous CADM1 expression patterns within resistant populations.

    • Live-Cell Imaging: Implement real-time visualization of CADM1 dynamics during drug exposure using fluorescent tagging and high-content microscopy.

    • Circulating Tumor Cell (CTC) Analysis: Evaluate CADM1 expression in CTCs as a potential liquid biopsy approach for monitoring developing resistance.

    • Spatial Transcriptomics/Proteomics: Apply spatially-resolved omics techniques to map CADM1 expression patterns within the tumor microenvironment and their relationship to treatment response.

Product Science Overview

Structure and Function

CADM1 is a type I transmembrane glycoprotein composed of three extracellular immunoglobulin-like domains, a single transmembrane domain, and a short cytoplasmic tail . The extracellular domains are responsible for mediating homophilic (CADM1-CADM1) and heterophilic (CADM1-CADM3, CADM1-NECTIN3) cell-cell adhesion . This adhesion is crucial for maintaining the structural integrity of tissues and facilitating communication between cells.

Biological Roles

CADM1 is involved in several biological processes, including:

  • Cell Recognition: CADM1 enables cells to recognize and adhere to each other, which is essential for tissue formation and maintenance .
  • Immune Response: CADM1 interacts with CRTAM (Class I-restricted T cell-associated molecule) to promote natural killer (NK) cell cytotoxicity and interferon-gamma (IFN-γ) secretion by CD8+ T cells . This interaction is vital for the immune system’s ability to target and destroy tumor cells.
  • Nervous System: In mast cells, CADM1 may mediate attachment to and communication with nerves, playing a role in the development and survival of mast cells .
Clinical Significance

CADM1 has been implicated in various diseases and conditions:

  • Cancer: CADM1 acts as a tumor suppressor in several cancers, including lung cancer, breast carcinoma, and prostate cancer . Its expression is often downregulated in these cancers, leading to increased tumor growth and metastasis.
  • Obesity: Genome-wide association studies have identified an association between body mass index (BMI) and loci near the CADM1 gene . Experimental studies have shown that loss of CADM1 can protect mice from obesity by promoting a negative energy balance and weight loss .
  • Anorexia Nervosa: A 2019 study revealed an association between CADM1 and anorexia nervosa, suggesting a potential role in the regulation of body weight and eating behavior .
Recombinant CADM1

Recombinant CADM1 is produced using recombinant DNA technology, where the CADM1 gene is cloned and expressed in suitable host cells, such as bacteria or mammalian cells. This allows for the production of large quantities of CADM1 protein for research and therapeutic purposes. Recombinant CADM1 is used in various applications, including:

  • Research: Studying the molecular mechanisms of cell adhesion and signaling pathways.
  • Therapeutics: Developing potential treatments for diseases where CADM1 is implicated, such as cancer and immune 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 2024 Thebiotek. All Rights Reserved.