C1QTNF6 Human

Complement C1q Tumor Necrosis Factor-Related Protein 6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Tissue Expression and Localization

C1QTNF6 exhibits tissue-specific expression, with notable activity in:

TissueExpression LevelCell TypeSource
PlacentaHighSyncytiotrophoblasts, endothelial cells
Adipose tissueModerateAdipocytes
EndometriumHighStromal cells
Colon/RectumModerateMucosal lymphoid cells

In cancer, overexpression is observed in 22 tumor types, including clear cell renal carcinoma (ccRCC), non-small cell lung cancer (NSCLC), and uterine carcinosarcoma .

Functional Roles in Cancer Biology

C1QTNF6 drives oncogenic processes through multiple mechanisms:

Cell Proliferation and Apoptosis

  • NSCLC: Overexpression promotes cell proliferation and reduces apoptosis via PI3K/Akt signaling .

  • Oral Squamous Cell Carcinoma (OSCC): Stimulates proliferation and inhibits apoptosis, linked to laminin receptor antagonism .

Tumor Microenvironment (TME) Modulation

PathwayAssociationClinical Implication
EMT (Epithelial-Mesenchymal Transition)Strong correlation with C1QTNF6 expression Enhances invasion and metastasis
AngiogenesisActivates VEGF-related pathways Promotes tumor vascularization
Immune CheckpointsCorrelates with PDCD1, CTLA4, and LAG3 Suppresses antitumor immunity

Immune Cell Infiltration

C1QTNF6 alters immune cell dynamics:

Immune CellCorrelationFunctional Impact
CD8+ T cellsNegative Reduced cytotoxic T-cell infiltration
MacrophagesPositive Promotes M2-like polarization
B cellsNegative Impaired humoral immunity

Diagnostic Utility

C1QTNF6 serves as a diagnostic marker in ccRCC, with high sensitivity and specificity for distinguishing tumor from normal tissue .

Drug Sensitivity and Resistance

Elevated C1QTNF6 levels correlate with reduced drug efficacy (high IC50 values) for 198 chemotherapeutic agents, including doxorubicin and cisplatin . This association highlights its role in multidrug resistance mechanisms.

Therapeutic Targeting and Future Directions

  1. Immune Checkpoint Inhibition: C1QTNF6’s correlation with PDCD1 and CTLA4 suggests potential synergy with anti-PD-1/PD-L1 therapies .

  2. EMT and Angiogenesis Inhibition: Targeting C1QTNF6 may suppress metastasis and tumor vascularization .

  3. Drug Sensitivity Optimization: Biomarker-driven stratification for personalized therapies .

Research Gaps and Challenges

  • Mechanistic Insights: Limited understanding of receptor interactions (e.g., laminin receptor in OSCC) .

  • Sex-Specific Roles: Higher expression in female placental and endometrial tissues warrants gender-specific studies .

  • Preclinical Validation: Most findings derive from bioinformatics; in vivo models are needed to confirm therapeutic potential .

Product Specs

Introduction
C1QTNF6, a type IV collagen, is found exclusively in basement membranes and plays a crucial role in embryonic development.
Description
Recombinant human C1QTNF6, produced in E. coli, is a non-glycosylated polypeptide chain consisting of 242 amino acids. With a molecular weight of 27 kDa, it includes an N-terminal His tag for purification. The amino acid sequence aligns with residues 29-259 of the UniProtKB/Swiss-Prot entry Q9BXI9. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The protein is supplied at a concentration of 0.23 mg/ml in a 30 mM acetate buffer with a pH of 4.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freeze at -20°C. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
CTRP6, C1QTNF6, ZACRP6, Complement C1q tumor necrosis factor-related protein 6.
Source
Escherichia Coli.
Amino Acid Sequence

MKHHHHHHAS TFDRAVASGC QRCCDSEDPL DPAHVSSASS SGRPHALPEI RPYINITILK GDKGDPGPMG LPGYMGREGP QGEPGPQGSK GDKGEMGSPG APCQKRFFAF SVGRKTALHS GEDFQTLLFE RVFVNLDGCF DMATGQFAAP LRGIYFFSLN VHSWNYKETY VHIMHNQKEA VILYAQPSER SIMQSQSVML DLAYGDRVWV RLFKRQRENA IYSNDFDTYI TFSGHLIKAEDD.

Q&A

What is C1QTNF6 and how is it characterized in normal human tissues?

C1QTNF6 (C1q and TNF-related protein 6) is a protein-coding gene that belongs to the C1QTNF family. In normal human tissues, C1QTNF6 shows variable expression patterns across different tissue types. Researchers typically characterize its expression using RNA sequencing data from databases like GTEx (Genotype-Tissue Expression), which provides baseline expression profiles across multiple tissue types. For comprehensive characterization, a combination of qPCR, western blotting, and immunohistochemistry should be employed to validate expression at both mRNA and protein levels .

How can researchers accurately measure C1QTNF6 expression in human samples?

To accurately measure C1QTNF6 expression, researchers should employ a multi-platform approach. RNA-sequencing provides comprehensive transcriptomic data, while RT-qPCR offers targeted validation. At the protein level, western blotting and immunohistochemistry (IHC) are standard methods. When analyzing tumor samples, it's critical to use paired tumor-normal tissues when possible, as demonstrated in the pan-cancer analysis methodologies. Researchers should be aware that comparative analyses between tumor and normal tissues often require integration of data from different repositories (such as TCGA for tumor samples and GTEx for normal tissue profiles) with appropriate batch correction and normalization procedures .

What methodological approaches are recommended for conducting pan-cancer analysis of C1QTNF6?

For pan-cancer analysis of C1QTNF6, researchers should follow a systematic approach:

  • Data acquisition: Utilize established databases like TCGA for tumor data and GTEx for normal tissue profiles, accessible through platforms like UCSC Xena.

  • Expression analysis: Compare expression levels between matched tumor and normal tissues using statistical methods like T-tests.

  • Mutation and copy number analysis: Examine genetic alterations using platforms like cBioportal.

  • Methylation analysis: Analyze methylation patterns using HM450 data types.

  • Survival analysis: Perform univariate and multivariate Cox regression analyses for multiple survival endpoints (OS, DSS, DFI, PFI).

  • Pathway analysis: Conduct GSEA (Gene Set Enrichment Analysis) to identify associated biological pathways.

  • Immune correlation: Analyze relationships with immune cell infiltration and immune-related genes.

This comprehensive approach enables thorough characterization of C1QTNF6's role across cancer types .

In which cancer types is C1QTNF6 most significantly overexpressed compared to normal tissues?

C1QTNF6 demonstrates significant overexpression in 22 distinct cancer types. The highest differential expression is observed in uterine carcinosarcoma (UCS), cholangiocarcinoma (CHOL), stomach adenocarcinoma (STAD), and diffuse large B-cell lymphoma (DLBC). When analyzing paired tumor-normal samples specifically, C1QTNF6 shows significant upregulation in 13 cancer types, with particularly strong overexpression in kidney renal papillary cell carcinoma (KIRP), head and neck squamous cell carcinoma (HNSC), liver hepatocellular carcinoma (LIHC), and esophageal carcinoma (ESCA). Notably, C1QTNF6 is significantly downregulated in kidney chromophobe (KICH), adrenocortical carcinoma (ACC), prostate adenocarcinoma (PRAD), and thyroid carcinoma (THCA) .

What mechanisms regulate C1QTNF6 expression in cancer cells?

C1QTNF6 expression in cancer cells is regulated through multiple mechanisms:

  • Copy Number Alterations (CNA): Strong positive correlations between C1QTNF6 expression and CNA have been observed in cholangiocarcinoma (CHOL), uterine carcinosarcoma (UCS), and pheochromocytoma and paraganglioma (PCPG), suggesting genetic amplification as a key regulatory mechanism.

  • Epigenetic regulation: Methylation analysis reveals significant correlations between C1QTNF6 expression and methylation patterns. Specifically, acute myeloid leukemia (LAML) shows positive correlation with methylation, while adrenocortical carcinoma (ACC) and uveal melanoma (UVM) demonstrate strong negative correlations, indicating cancer-specific epigenetic regulation.

  • Transcriptional regulation: Associations with various signaling pathways, including TGF-β, NF-κB, and IL2-STAT5, suggest complex transcriptional control mechanisms.

These diverse regulatory mechanisms contribute to the heterogeneous expression patterns observed across different cancer types .

How does C1QTNF6 genetic alteration frequency vary across different cancer types?

The genetic alteration frequency of C1QTNF6 varies considerably across cancer types. Uterine carcinosarcoma (UCS) exhibits the highest alteration frequency, with amplification mutations being the predominant type. Other cancers show varying frequencies and types of alterations. When examining the relationship between C1QTNF6 expression and copy number alterations (CNA), strong positive correlations are observed in cholangiocarcinoma (CHOL), uterine carcinosarcoma (UCS), and pheochromocytoma and paraganglioma (PCPG). This suggests that in these cancers, copy number gains significantly contribute to increased C1QTNF6 expression. Researchers investigating C1QTNF6 in specific cancer types should first assess the prevalence and type of genetic alterations to inform experimental design and interpretation of expression data .

What is the prognostic significance of C1QTNF6 in different cancer types?

C1QTNF6 serves as an independent prognostic indicator across multiple cancer types. Comprehensive survival analyses using univariate Cox regression revealed significant associations with:

Multivariate Cox regression analyses confirmed C1QTNF6 as an independent prognostic factor after adjustment for other clinical characteristics in multiple cancers, including KIRC, LUAD, LGG, KIRP, ACC, UVM, MESO, LIHC, and HNSC. In most cancers, elevated C1QTNF6 expression is associated with unfavorable outcomes (hazard ratio > 1), highlighting its potential as a risk biomarker .

How can researchers validate C1QTNF6 as a prognostic biomarker in their specific cancer cohorts?

To validate C1QTNF6 as a prognostic biomarker in specific cancer cohorts, researchers should follow these methodological steps:

  • Cohort Selection: Include both tumor and adjacent normal tissues with comprehensive clinical follow-up data encompassing multiple survival endpoints (OS, DSS, DFI, PFI).

  • Expression Analysis: Quantify C1QTNF6 expression using both RNA-seq and protein-level methods (western blot, IHC) to ensure robust detection.

  • Statistical Validation:

    • Perform univariate Cox regression to assess basic prognostic significance

    • Conduct multivariate Cox regression adjusting for established clinical parameters (age, stage, grade)

    • Calculate hazard ratios with confidence intervals

    • Generate Kaplan-Meier survival curves stratifying patients by C1QTNF6 expression levels

  • Subgroup Analysis: Evaluate prognostic value across different patient subgroups based on clinicopathological features.

  • External Validation: Confirm findings in independent cohorts to establish reproducibility.

This comprehensive approach can establish C1QTNF6 as a clinically relevant biomarker while identifying cancer types where it provides the most significant prognostic information .

How does C1QTNF6 expression correlate with response to common cancer therapies?

C1QTNF6 expression demonstrates significant correlations with drug sensitivity across multiple cancer treatments. Analysis using the GDSC2 database (containing 198 drugs) revealed that higher C1QTNF6 expression predicts higher IC50 values for most drugs, indicating potential drug resistance mechanisms. This relationship was systematically evaluated by:

  • Spearman correlation analysis between C1QTNF6 expression and drug sensitivity

  • Comparison of drug sensitivity between low and high C1QTNF6 expression groups

  • Analysis of differences in response to commonly used anticancer drugs using the Kruskal-Wallis rank-sum test

The findings demonstrate that C1QTNF6 overexpression may contribute to treatment resistance in multiple cancer types, potentially through mechanisms related to epithelial-mesenchymal transition (EMT), which has been widely linked to drug resistance. This suggests that C1QTNF6 expression levels could serve as a predictive biomarker for treatment response and that targeting C1QTNF6-related pathways might enhance therapeutic efficacy .

Which biological pathways are most significantly associated with C1QTNF6 expression?

C1QTNF6 expression shows strong correlations with multiple critical cancer-related pathways. The most significant associations include:

  • Epithelial-Mesenchymal Transition (EMT): Strong positive correlation across multiple cancer types, suggesting C1QTNF6's involvement in cancer cell invasion, migration, and metastasis.

  • Tumor Microenvironment (TME) Pathways: Significant associations with base excision repair, mismatch repair, and angiogenesis pathways.

  • Immune-Related Pathways: Correlations with immune checkpoint regulation, CD8 T effector pathways, and various immune system processes including cytokine pathways, antigen processing/presentation, neutrophil degranulation, and interleukin immunomodulatory responses.

  • Signaling Pathways: Associations with TGF-β signaling, TNF-α signaling via NF-κB, IL2-STAT5 signaling, Notch pathway, and hypoxia response.

These diverse pathway associations suggest C1QTNF6 functions at the intersection of tumor progression, immune regulation, and microenvironment modulation. Gene Set Enrichment Analysis (GSEA) confirmed these associations across multiple cancer types, with immune-related pathways particularly prominent in uterine carcinosarcoma (UCS) .

What is the relationship between C1QTNF6 and tumor microenvironment components?

C1QTNF6 shows significant associations with tumor microenvironment (TME) components across multiple cancers. Key relationships include:

  • Stromal and Immune Scores: C1QTNF6 expression correlates with stromal scores, estimate scores, and immune scores, indicating potential involvement in regulating non-tumor components of the TME.

  • Immune Cell Infiltration: C1QTNF6 expression demonstrates negative correlations with several immune cell populations, including B cells, neutrophils, and CD8+ T cells, while showing positive correlations with monocytes and macrophages. This suggests a potential immunomodulatory role that may contribute to immune evasion in tumors with high C1QTNF6 expression.

  • Immune-Related Pathways: Gene Set Enrichment Analysis (GSEA) identified significant associations between C1QTNF6 and immune system processes, cytokine signaling, antigen processing/presentation, neutrophil degranulation, and interleukin responses.

These relationships indicate that C1QTNF6 plays a critical role in shaping the immune landscape within the tumor microenvironment, potentially contributing to immune escape mechanisms that facilitate tumor progression .

How does C1QTNF6 influence cancer hallmark processes like EMT and angiogenesis?

C1QTNF6 demonstrates strong positive correlations with critical cancer hallmark processes, most notably epithelial-mesenchymal transition (EMT) and angiogenesis. The mechanisms through which C1QTNF6 influences these processes include:

  • Epithelial-Mesenchymal Transition (EMT):

    • C1QTNF6 shows significant correlation with EMT pathway activation

    • EMT enables cancer cells to acquire enhanced proliferative, invasive, and migratory capacities

    • EMT is linked to drug resistance, which aligns with findings that high C1QTNF6 expression predicts poorer drug responses

    • This relationship may explain why C1QTNF6 overexpression correlates with poor prognosis in multiple cancer types

  • Angiogenesis:

    • C1QTNF6 positively correlates with angiogenesis pathways

    • Angiogenesis is essential for tumor growth, proliferation, and invasion

    • This correlation suggests C1QTNF6 may play a role in promoting blood vessel formation within tumors

    • Understanding this relationship could inform development of novel anti-angiogenic therapeutic approaches

  • Additional Hallmark Pathways:

    • Significant associations with apical junction formation

    • Correlations with TGF-β signaling pathway, hypoxia response, and Notch signaling

    • Connections to immune-related pathways including TNF-α signaling via NF-κB and IL2-STAT5 signaling

These relationships position C1QTNF6 at the intersection of multiple cancer hallmark processes, providing potential explanations for its prognostic significance and suggesting possible mechanisms through which it influences tumor progression .

What experimental approaches are most effective for studying C1QTNF6 function in cancer models?

For comprehensive investigation of C1QTNF6 function in cancer, researchers should implement a multi-faceted experimental approach:

  • Expression Modulation:

    • CRISPR-Cas9 knockout or knockdown via shRNA/siRNA for loss-of-function studies

    • Lentiviral/retroviral overexpression systems for gain-of-function experiments

    • Inducible expression systems to study temporal effects

  • In Vitro Functional Assays:

    • Proliferation assays (MTT, BrdU incorporation)

    • Migration/invasion assays (transwell, wound healing)

    • Drug sensitivity assays with IC50 determination

    • Co-culture systems with immune cells to assess immunomodulatory effects

  • In Vivo Models:

    • Xenograft models with C1QTNF6-modulated cell lines

    • Patient-derived xenografts

    • Genetically engineered mouse models when appropriate

  • Molecular Mechanism Investigation:

    • RNA-seq and proteomic analysis to identify downstream targets

    • ChIP-seq to evaluate transcriptional regulation

    • Co-immunoprecipitation to identify protein interaction partners

    • Pathway inhibitors to dissect signaling connections

  • Clinical Correlation:

    • Tissue microarrays for protein expression validation

    • Analysis of mutation, methylation, and CNA data in patient cohorts

    • Integration with treatment response data

This comprehensive approach will enable researchers to establish causative relationships between C1QTNF6 expression and cancer phenotypes while elucidating the underlying molecular mechanisms .

How can researchers investigate the potential of C1QTNF6 as a therapeutic target?

To investigate C1QTNF6 as a therapeutic target, researchers should pursue a systematic drug development pathway:

  • Target Validation:

    • Confirm oncogenic role through knockout/knockdown studies in multiple cancer models

    • Establish association with clinically relevant phenotypes (proliferation, metastasis, drug resistance)

    • Validate in patient-derived samples and animal models

    • Determine cancer types most likely to benefit from C1QTNF6 targeting

  • Therapeutic Strategy Development:

    • Direct targeting: Develop blocking antibodies or small molecule inhibitors

    • Indirect targeting: Identify and inhibit downstream effectors or upstream regulators

    • Combination approaches: Test with standard chemotherapies, immunotherapies, or targeted agents based on pathway interactions

  • Predictive Biomarker Identification:

    • Establish expression thresholds that predict response

    • Identify genetic or molecular features that enhance sensitivity

    • Develop companion diagnostics to guide patient selection

  • Resistance Mechanism Investigation:

    • Study adaptive responses to C1QTNF6 inhibition

    • Identify bypass pathways that confer resistance

    • Develop rational combination strategies to overcome resistance

  • Translational Studies:

    • Evaluate pharmacodynamic biomarkers

    • Conduct proof-of-concept studies in patient-derived xenografts

    • Design early-phase clinical trials with biological endpoints

Given C1QTNF6's association with drug resistance and its links to critical cancer hallmarks like EMT and angiogenesis, targeting this protein could potentially overcome treatment resistance mechanisms in multiple cancer types .

What are the challenges in translating C1QTNF6 research findings to clinical applications?

Translating C1QTNF6 research findings to clinical applications faces several significant challenges:

  • Biological Complexity:

    • Context-dependent expression patterns across different cancer types

    • Dual role in certain cancers (downregulated in some, upregulated in others)

    • Complex interplay with multiple signaling pathways and immune components

    • Potential redundancy with other C1QTNF family members

  • Methodological Barriers:

    • Need for standardized detection methods with appropriate sensitivity and specificity

    • Challenges in establishing clinically relevant cutoff values for "high" versus "low" expression

    • Requirement for prospective validation in diverse clinical cohorts

    • Limited availability of well-characterized antibodies for protein detection

  • Clinical Translation Challenges:

    • Necessity to develop cancer-specific prognostic models integrating C1QTNF6 with other biomarkers

    • Need for prospective clinical trials to validate prognostic and predictive value

    • Potential challenges in drug development (target accessibility, specificity)

    • Determining optimal timing for therapeutic intervention

  • Practical Implementation Issues:

    • Integration into existing clinical workflows

    • Cost-effectiveness considerations for routine testing

    • Educational requirements for clinicians

    • Regulatory approval pathways

Addressing these challenges requires interdisciplinary collaboration between basic scientists, translational researchers, clinicians, and industry partners to validate C1QTNF6's clinical utility and develop effective targeting strategies .

What statistical approaches are most appropriate for analyzing C1QTNF6 expression data in clinical cohorts?

For rigorous analysis of C1QTNF6 expression in clinical cohorts, researchers should employ the following statistical approaches:

  • Expression Comparison Analysis:

    • T-tests for comparing tumor vs. normal expression (paired when possible)

    • ANOVA with post-hoc tests for multi-group comparisons

    • Non-parametric alternatives (Mann-Whitney U, Kruskal-Wallis) when data violates normality assumptions

  • Correlation Analysis:

    • Pearson's correlation for normally distributed continuous variables (e.g., with CNA, methylation data)

    • Spearman's correlation for non-parametric relationships (e.g., with drug sensitivity, immune scores)

  • Survival Analysis:

    • Univariate Cox regression to assess basic prognostic value

    • Multivariate Cox regression adjusting for established clinical parameters

    • Kaplan-Meier method with log-rank tests for visualizing survival differences

    • Analysis of multiple survival endpoints (OS, DSS, DFI, PFI) for comprehensive evaluation

  • Machine Learning Approaches:

    • Random forest or LASSO regression for feature selection in multi-marker panels

    • Cross-validation to assess model robustness

    • Independent validation cohorts to confirm findings

  • Multiple Testing Correction:

    • Bonferroni or False Discovery Rate methods to control for type I errors in multi-cancer analyses

This comprehensive statistical framework ensures reliable interpretation of C1QTNF6's clinical significance while minimizing bias and confounding effects .

How can researchers address potential confounding factors when studying C1QTNF6 in patient samples?

To address potential confounding factors when studying C1QTNF6 in patient samples, researchers should implement the following methodological approaches:

  • Sample Selection and Characterization:

    • Match samples for key demographic variables (age, sex, ethnicity)

    • Stratify by tumor stage, grade, and molecular subtypes

    • Collect comprehensive clinical data including treatment history

    • Use paired tumor-normal samples when possible to control for individual variability

  • Analytical Controls:

    • Include reference genes/proteins with stable expression for normalization

    • Implement batch correction methods for multi-center datasets

    • Use technical replicates to assess measurement variability

    • Include positive and negative controls in all experimental platforms

  • Statistical Handling of Confounders:

    • Perform multivariate analyses adjusting for known prognostic factors

    • Use propensity score matching for treatment effect studies

    • Implement stratified analyses for key subgroups

    • Conduct sensitivity analyses to assess robustness of findings

  • Data Integration Approaches:

    • Integrate multi-omics data (transcriptomics, proteomics, methylation)

    • Cross-reference with multiple databases (TCGA, GTEx, GDSC)

    • Validate findings across different analytical platforms

    • Apply causal inference methods when appropriate

  • Validation Strategies:

    • Use independent cohorts for external validation

    • Implement different detection methodologies to confirm findings

    • Consider temporal validation (samples collected at different time points)

    • Conduct meta-analyses when multiple datasets are available

What are the key considerations for designing functional studies to elucidate C1QTNF6's role in cancer progression?

When designing functional studies to elucidate C1QTNF6's role in cancer progression, researchers should consider the following key factors:

  • Model Selection:

    • Choose cell lines with varying baseline C1QTNF6 expression levels

    • Prioritize cancer types where C1QTNF6 shows strong prognostic significance

    • Include normal cell counterparts as controls

    • Consider patient-derived models for greater clinical relevance

  • Expression Modulation Strategy:

    • For knockdown: evaluate efficiency and specificity of different approaches (siRNA, shRNA, CRISPR)

    • For overexpression: consider physiologically relevant expression levels

    • Use inducible systems to study temporal effects

    • Implement rescue experiments to confirm specificity

  • Phenotypic Assays:

    • Align functional assays with C1QTNF6's known associations:

      • Proliferation and survival assays (connected to prognostic significance)

      • Migration/invasion assays (related to EMT correlation)

      • Angiogenesis models (based on pathway associations)

      • Drug response assays (given correlation with treatment resistance)

      • Immune co-culture systems (based on immune microenvironment associations)

  • Mechanistic Investigations:

    • Pathway analysis focusing on EMT, angiogenesis, and immune modulation

    • Protein-protein interaction studies to identify binding partners

    • Transcriptional profiling to identify downstream targets

    • Upstream regulatory mechanism analysis (methylation, transcription factors)

  • In Vivo Validation:

    • Design xenograft studies with appropriate endpoints based on in vitro findings

    • Consider orthotopic models for microenvironment interactions

    • Include therapeutic intervention studies when relevant

    • Monitor immune infiltration in immunocompetent models

  • Translational Connections:

    • Validate key findings in patient samples

    • Correlate functional observations with clinical outcomes

    • Develop biomarker assays based on mechanism insights

This comprehensive approach will establish causative relationships between C1QTNF6 and cancer phenotypes while elucidating the underlying molecular mechanisms that could be therapeutically targeted .

What are promising research directions for understanding C1QTNF6's role in immune evasion mechanisms?

Given C1QTNF6's significant correlations with immune components, several promising research directions could elucidate its role in immune evasion:

  • Immune Cell Interaction Studies:

    • Co-culture experiments between C1QTNF6-manipulated cancer cells and various immune populations

    • Analysis of how C1QTNF6 affects T cell activation, proliferation, and cytokine production

    • Investigation of impact on antigen presentation and recognition

    • Examination of effects on NK cell cytotoxicity

  • Checkpoint Regulation Mechanisms:

    • Analysis of correlation between C1QTNF6 and established immune checkpoint molecules (PD-1, PD-L1, CTLA-4)

    • Investigation of potential direct or indirect regulation of checkpoint expression

    • Assessment of synergy between C1QTNF6 inhibition and checkpoint blockade

  • Cytokine/Chemokine Network Analysis:

    • Comprehensive profiling of cytokine/chemokine production in C1QTNF6-high vs. low tumors

    • Mechanistic studies on how C1QTNF6 modulates inflammatory signaling

    • Investigation of impact on chemotactic gradients affecting immune cell recruitment

  • Tumor Microenvironment Modulation:

    • Examination of how C1QTNF6 affects the polarization of tumor-associated macrophages

    • Analysis of impact on myeloid-derived suppressor cell recruitment and function

    • Investigation of effects on regulatory T cell induction

  • Integration with Immunotherapy Response:

    • Retrospective analysis of C1QTNF6 expression in immunotherapy responders vs. non-responders

    • Development of predictive models incorporating C1QTNF6 for immunotherapy efficacy

    • Testing combination approaches targeting C1QTNF6 alongside immunotherapies

These research directions would provide crucial insights into C1QTNF6's role in immune evasion, potentially leading to novel immunotherapeutic strategies for cancers with high C1QTNF6 expression .

How might single-cell analysis technologies advance our understanding of C1QTNF6 function?

Single-cell analysis technologies offer transformative potential for advancing our understanding of C1QTNF6 function in cancer:

  • Cellular Heterogeneity Characterization:

    • Single-cell RNA sequencing (scRNA-seq) to identify specific cell populations expressing C1QTNF6

    • Analysis of expression heterogeneity within tumors and across patients

    • Identification of rare cell populations with distinctive C1QTNF6 expression patterns

    • Characterization of co-expression patterns with other cancer-related genes

  • Spatial Context Integration:

    • Spatial transcriptomics to map C1QTNF6 expression within the tumor architecture

    • Investigation of expression gradients relative to vascular structures, immune infiltrates, and stromal boundaries

    • Multiplex immunofluorescence to visualize C1QTNF6 protein localization alongside immune markers

    • Correlation of spatial patterns with local microenvironmental features

  • Functional State Analysis:

    • CITE-seq to simultaneously profile surface protein markers and transcriptome

    • Single-cell ATAC-seq to characterize chromatin accessibility in C1QTNF6-expressing cells

    • Integration of multi-omic data to define functional cell states associated with C1QTNF6 expression

    • Trajectory analysis to map cellular transitions related to C1QTNF6 activation

  • Microenvironmental Interactions:

    • Single-cell TCR/BCR sequencing to characterize immune repertoire in relation to C1QTNF6 expression

    • Analysis of ligand-receptor interactions between C1QTNF6-expressing cells and surrounding cells

    • Characterization of intercellular communication networks in high vs. low C1QTNF6 environments

    • Single-cell secretome analysis to identify paracrine signaling patterns

  • Therapeutic Response Monitoring:

    • Single-cell profiling before and after therapeutic interventions

    • Identification of resistant cell populations and their relationship to C1QTNF6 expression

    • Characterization of treatment-induced changes in C1QTNF6-associated cellular programs

These advanced technologies would provide unprecedented resolution in understanding C1QTNF6's cellular context, regulation, and functional impact within the complex tumor ecosystem .

What potential exists for developing C1QTNF6-targeted therapeutics across different cancer types?

The development of C1QTNF6-targeted therapeutics offers promising potential across multiple cancer types, with several strategic approaches:

  • Direct Targeting Strategies:

    • Monoclonal antibodies against C1QTNF6 protein

    • Small molecule inhibitors targeting functional domains

    • Antisense oligonucleotides or siRNA for expression knockdown

    • Proteolysis-targeting chimeras (PROTACs) for targeted degradation

  • Cancer Type Prioritization:

    • Focus on cancers with highest C1QTNF6 overexpression (UCS, CHOL, STAD, DLBC)

    • Prioritize malignancies where C1QTNF6 shows strongest prognostic significance (KIRC, LUAD, LGG)

    • Target cancers with established drug resistance challenges

    • Consider cancers with limited current therapeutic options

  • Combination Therapy Approaches:

    • Integration with existing chemotherapies to overcome resistance

    • Combination with immune checkpoint inhibitors based on immune correlations

    • Pairing with anti-angiogenic agents given pathway associations

    • Development of dual-targeting approaches addressing C1QTNF6 and EMT pathways

  • Precision Medicine Applications:

    • Development of companion diagnostics for patient selection

    • Identification of biomarkers predicting response to C1QTNF6 targeting

    • Creation of C1QTNF6-based molecular subtypes to guide therapy

    • Implementation of adaptive trial designs based on C1QTNF6 expression

  • Novel Delivery Approaches:

    • Nanoparticle-based delivery systems for tumor-specific targeting

    • Antibody-drug conjugates incorporating C1QTNF6 antibodies

    • Cell-based therapies engineered to recognize C1QTNF6-expressing cells

    • Local delivery strategies for cancers with accessible anatomical locations

The strong associations between C1QTNF6 and drug resistance, coupled with its connections to EMT and immune modulation, position it as a promising therapeutic target with potential to address significant unmet needs in cancer treatment .

What are the key challenges in developing reliable antibodies and detection methods for C1QTNF6?

Developing reliable antibodies and detection methods for C1QTNF6 presents several technical challenges that researchers must address:

  • Antibody Development Challenges:

    • Protein structure complexity and potential post-translational modifications

    • Cross-reactivity with other C1QTNF family members due to sequence homology

    • Limited immunogenicity of certain epitopes

    • Potential conformational differences between recombinant and native protein

  • Validation Requirements:

    • Comprehensive validation across multiple detection platforms (western blot, IHC, flow cytometry)

    • Verification using negative controls (knockout/knockdown samples)

    • Comparison with orthogonal detection methods (RNA-seq, qPCR)

    • Testing across diverse sample types (fresh tissue, FFPE, cell lines)

  • Technical Optimization Considerations:

    • Antigen retrieval optimization for formalin-fixed tissues

    • Titration to determine optimal antibody concentrations

    • Signal amplification strategies for low-expression contexts

    • Blocking protocol refinement to minimize background

  • Reproducibility Concerns:

    • Lot-to-lot variability in commercial antibodies

    • Standardization of staining protocols across laboratories

    • Development of quantitative scoring systems

    • Implementation of appropriate controls for multi-center studies

  • Emerging Technology Integration:

    • Adaptation for multiplexed immunofluorescence

    • Optimization for mass cytometry applications

    • Development of proximity ligation assays for protein interaction studies

    • Creation of aptamer-based detection alternatives

Addressing these challenges is essential for generating reliable data on C1QTNF6 expression and localization, which forms the foundation for all subsequent functional and clinical studies .

How should researchers approach the analysis of C1QTNF6 in the context of tumor heterogeneity?

Tumor heterogeneity presents significant challenges for C1QTNF6 analysis, requiring specialized methodological approaches:

  • Sampling Strategies:

    • Multi-region sampling to capture intratumoral heterogeneity

    • Inclusion of primary and metastatic sites when available

    • Temporal sampling to assess expression changes during disease progression

    • Comparison of treatment-naïve and post-treatment samples

  • Single-Cell Resolution Techniques:

    • Single-cell RNA sequencing to identify distinct cell populations expressing C1QTNF6

    • Single-cell proteomics to characterize protein-level heterogeneity

    • Spatial transcriptomics or multiplexed immunofluorescence to map expression in tissue context

    • Integration of multi-omic data at single-cell level

  • Computational Approaches:

    • Deconvolution algorithms for bulk RNA-seq data to estimate cell type-specific expression

    • Clustering methods to identify patient subgroups with distinct C1QTNF6 expression patterns

    • Trajectory analysis to model evolution of C1QTNF6-expressing clones

    • Network analysis to identify context-dependent co-expression patterns

  • Experimental Models:

    • Patient-derived xenografts that maintain tumor heterogeneity

    • Organoid cultures from different tumor regions

    • Co-culture systems incorporating multiple cell types

    • In vivo lineage tracing to track C1QTNF6-expressing populations

  • Clinical Translation Considerations:

    • Development of heterogeneity-aware biomarker strategies

    • Establishment of sampling guidelines for diagnostic testing

    • Design of therapeutic approaches addressing heterogeneous expression

    • Implementation of adaptive monitoring during treatment

These approaches enable researchers to characterize C1QTNF6 expression patterns within heterogeneous tumors, providing crucial insights for translation to clinical applications that account for this biological complexity .

What are the most important considerations when interpreting contradictory findings about C1QTNF6 across different cancer studies?

When interpreting contradictory findings about C1QTNF6 across different cancer studies, researchers should consider several critical factors:

  • Biological Context Differences:

    • Cancer-type specificity (C1QTNF6 shows divergent expression patterns across cancer types)

    • Stage-dependent effects (expression and function may vary with disease progression)

    • Microenvironmental context (influence of surrounding tissue and immune components)

    • Molecular subtype variations (effects may differ across established molecular classifications)

  • Methodological Variations:

    • Detection method differences (RNA vs. protein, different antibody clones)

    • Sample processing variations (fresh vs. fixed tissue, extraction protocols)

    • Quantification approaches (continuous vs. categorical analysis)

    • Reference/control selection disparities

  • Statistical Considerations:

    • Sample size limitations affecting power to detect associations

    • Multiple testing issues in high-dimensional studies

    • Confounding factors not adequately addressed

    • Endpoint definition differences (OS vs. PFS vs. treatment response)

  • Study Design Factors:

    • Retrospective vs. prospective design

    • Population differences (geographic, demographic, treatment strategies)

    • Selection bias in cohort composition

    • Follow-up duration disparities

  • Analytical Framework:

    • Perform systematic reviews incorporating quality assessment

    • Conduct meta-analyses when appropriate

    • Consider Bayesian approaches to integrate prior knowledge

    • Implement triangulation of evidence across methodologies

  • Biological Plausibility:

    • Assess consistency with known molecular mechanisms

    • Evaluate coherence with established cancer biology principles

    • Consider species differences when comparing human and model organism studies

    • Examine context-dependent signaling networks

Recognizing these factors enables researchers to synthesize seemingly contradictory findings into a more nuanced understanding of C1QTNF6's context-dependent roles in cancer biology .

Product Science Overview

Introduction

Complement C1q Tumor Necrosis Factor-Related Protein 6 (C1QTNF6) is a member of the C1q/TNF-related protein (CTRP) family. This family consists of secreted proteins that share structural similarities with the complement protein C1q and the tumor necrosis factor (TNF) superfamily. CTRPs are known for their roles in various physiological processes, including metabolism, inflammation, and immune response .

Structure

C1QTNF6, like other CTRPs, is composed of several distinct domains:

  1. N-terminal signal peptide: This domain is responsible for directing the protein to the secretory pathway.
  2. Short variable domain: This region varies among different CTRPs and contributes to their unique functions.
  3. Collagenous domain: This domain is characterized by a series of Gly-X-Y repeats, where X and Y are often proline and hydroxyproline, respectively. It is involved in the formation of higher-order structures.
  4. C-terminal globular C1q domain: This domain is homologous to the globular domain of the complement protein C1q and is crucial for the protein’s biological activity .
Function

C1QTNF6 has been implicated in various biological processes, including:

  • Metabolism: C1QTNF6 plays a role in regulating glucose and lipid metabolism. It has been shown to enhance insulin sensitivity and promote fatty acid oxidation .
  • Inflammation: C1QTNF6 exhibits anti-inflammatory properties by modulating the activity of immune cells and reducing the production of pro-inflammatory cytokines .
  • Cellular signaling: C1QTNF6 activates several signaling pathways, including the AMP-activated protein kinase (AMPK) and AKT pathways, which are involved in maintaining cellular energy homeostasis and promoting cell survival .
Recombinant Human C1QTNF6

Recombinant human C1QTNF6 is produced using recombinant DNA technology, which involves inserting the gene encoding C1QTNF6 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 Therapeutic Potential

C1QTNF6 has garnered significant interest in the scientific community due to its potential therapeutic applications. Studies have shown that C1QTNF6 can:

  • Improve metabolic health: By enhancing insulin sensitivity and promoting lipid metabolism, C1QTNF6 may help in the management of metabolic disorders such as obesity, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD) .
  • Reduce inflammation: The anti-inflammatory properties of C1QTNF6 make it a promising candidate for the treatment of inflammatory diseases, including autoimmune disorders and cardiovascular diseases .
  • Support cancer therapy: Although C1QTNF6 does not directly affect tumor growth, it has been shown to reduce hypovascular tumor necrosis areas, potentially improving the efficacy of cancer treatments .

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.