C1QTNF7 Human

Complement C1q Tumor Necrosis Factor-Related Protein 7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Production and Biochemical Properties

C1QTNF7 Human is typically produced via recombinant expression in E. coli, yielding a lyophilized powder suitable for laboratory use . Key biochemical details:

  • Purity: >95% as confirmed by SDS-PAGE .

  • Solubility: Requires reconstitution in 0.1M acetate buffer (pH 4) for optimal solubility; higher pH may reduce solubility .

  • Stability: Stable at -20°C for long-term storage; reconstituted solutions remain viable at 4°C for ≤2 weeks .

Functional Roles and Interactions

C1QTNF7 Human exhibits diverse biological functions:

Collagen Trimerization and Matrix Organization

  • Collagen type IV specificity: Binds to basement membrane collagen, facilitating extracellular matrix remodeling .

  • Structural support: Part of collagen trimer complexes critical for tissue integrity .

Cancer Biology and Prognosis

High expression of C1QTNF7 correlates with survival outcomes in multiple cancers:

Table 2: C1QTNF7 Expression and Cancer Prognosis

Cancer TypeExpression PatternSurvival Correlation (p<0.001)Source
BreastHigh in tumor microenvironmentUnfavorable prognosis
ColorectalElevated in metastatic tissuesUnfavorable prognosis
ProstateVariable expressionContext-dependent prognosis

Data from The Cancer Genome Atlas (TCGA) and immunohistochemistry studies highlight its role in tumor progression .

Functional Associations

C1QTNF7 interacts with 2,048 biological entities across molecular profiles, diseases, and pathways . Key associations include:

  • Molecular interactions: Collagen-binding proteins, TNF superfamily members .

  • Disease links: Inflammatory disorders, cardiovascular diseases .

Genomic Context and Evolutionary Conservation

  • Genomic location: Chromosome 4p15.32, spanning exons with conserved sequences across species .

  • Orthologs: Evolutionary conservation in rat (C1qtnf7) and mouse models .

Research Applications and Future Directions

C1QTNF7 Human serves as a critical reagent in:

  • Cancer research: Biomarker discovery for prognosis and therapeutic targeting .

  • Structural biology: Studying collagen interactions and extracellular matrix dynamics .

Table 3: Functional Datasets and Tools

Dataset/ToolApplicationSource
Human Protein AtlasTissue-specific expression maps
HarmonizomePathway and interaction networks
DepMap CRISPRGene dependency analysis

Product Specs

Introduction
C1QTNF7, a member of the C1Q and TNF superfamily, is a collagen type-IV protein predominantly found in basement membranes.
Description
Recombinant Human C1QTNF7, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 283 amino acids, with a molecular weight of 30.22 kDa. Notably, a 10 amino acid His-tag is present at the N-terminus. The amino acid sequence of this C1QTNF7 variant perfectly aligns with amino acids 17-289 of the UniProtKB/Swiss-Prot entry Q9BXJ2. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
The product appears as a white, lyophilized (freeze-dried) powder after filtration.
Formulation
For lyophilization, Human C1QTNF7 undergoes a 0.4µm filtration process. The lyophilization buffer consists of 30mM Acetate Buffer (pH 4) and 5% (w/v) trehalose, with an initial protein concentration of 0.5mg/ml.
Solubility
To prepare a working stock solution of approximately 0.5mg/mL, reconstitute the lyophilized pellet by adding 0.1M Acetate buffer (pH 4) and ensure complete dissolution. If a higher pH value is required, dilute the solution significantly with an appropriate buffer to a final concentration of 10µg/ml. High antigen concentrations may limit solubility. This product is not sterile; filter it through a sterile filter before cell culture applications.
Stability
Lyophilized protein should be stored at -20°C. After reconstitution, aliquot the product to minimize freeze/thaw cycles. While the reconstituted protein exhibits stability at 4°C for a short duration (up to two weeks with no observable changes), long-term storage at this temperature is not recommended.
Purity
Purity levels exceed 95% as determined by SDS-PAGE analysis.
Synonyms
CTRP7, C1QTNF7, ZACRP7, Complement C1q tumor necrosis factor-related protein 7.
Source
Escherichia Coli.
Amino Acid Sequence
MKHHHHHHAS QPRGNQLKGE NYSPRYICSI PGLPGPPGPP GANGSPGPHG RIGLPGRDGR DGRKGEKGEK GTAGLRGKTG PLGLAGEKGD QGETGKKGPI GPEGEKGEVG PIGPPGPKGD RGEQGDPGLP GVCRCGSIVL KSAFSVGITT SYPEERLPII FNKVLFNEGE HYNPATGKFI CAFPGIYYFS YDITLANKHL AIGLVHNGQY RIKTFDANTG NHDVASGSTV IYLQPEDEVW LEIFFTDQNG LFSDPGWADS LFSGFLLYVD TDYLDSISEDDEL.

Q&A

What is C1QTNF7 and what is its structure?

C1QTNF7 (Complement C1q Tumor Necrosis Factor-Related Protein 7) is a non-glycosylated polypeptide chain containing 283 amino acids with a molecular mass of 30.22 kDa. The protein's amino acid sequence is identical to UniProtKB/Swiss-Prot entry Q9BXJ2 amino acids 17-289. When produced as a recombinant protein in E.Coli, it typically contains an additional 10 amino acid His tag at the N-terminus to facilitate purification processes. C1QTNF7 belongs to the C1q/TNF family of proteins, which commonly feature a globular C1q domain at the C-terminus and a collagen-like domain near the N-terminus .

What is the tissue expression profile of C1QTNF7?

C1QTNF7 demonstrates a distinct tissue expression pattern across multiple human tissues. Based on protein atlas data, C1QTNF7 shows varying expression levels across multiple tissue types including adipose tissue, adrenal gland, brain regions (amygdala, basal ganglia, cerebellum, cerebral cortex), cardiovascular tissues (heart muscle), reproductive tissues (endometrium, fallopian tube, ovary, placenta), and other major organs (kidney, liver, lung) . Understanding this expression profile is crucial for contextualizing research findings and determining physiologically relevant experimental models for C1QTNF7 studies.

How should C1QTNF7 recombinant protein be stored and handled?

For optimal research outcomes, C1QTNF7 recombinant protein requires specific handling protocols. The lyophilized protein should be stored at -20°C until use. After reconstitution, it is crucial to aliquot the protein to avoid repeated freezing/thawing cycles which can compromise protein integrity and activity. Reconstituted protein can be stored at 4°C for a limited period of time; research indicates it shows no significant changes after two weeks at 4°C. For shipping and initial handling, the protein can be maintained at room temperature before proper storage . Researchers should document storage conditions and reconstitution dates in laboratory notebooks to ensure experimental reproducibility.

What are appropriate experimental designs for studying C1QTNF7 function?

When designing experiments to study C1QTNF7 function, researchers should consider both true experimental and quasi-experimental approaches depending on their research questions and constraints. True experimental designs with random assignment provide the strongest evidence for causal relationships but may not always be feasible. For in vitro studies, randomized controlled designs with appropriate cell lines that naturally express C1QTNF7 or knockout/overexpression models would be optimal. For in vivo or clinical studies, quasi-experimental designs may be necessary due to ethical or practical limitations .
A quasi-experimental approach might involve comparing naturally occurring groups with different C1QTNF7 expression levels or utilizing regression discontinuity designs around clinical thresholds. In either approach, controlling for confounding variables is essential, particularly when working with patient samples or complex tissue systems. Researchers should clearly document their experimental design rationale, control strategies, and statistical approaches to address the inherent limitations of their chosen design .

How can I validate antibodies for C1QTNF7 research?

Antibody validation is critical for reliable C1QTNF7 research. A multi-tier validation approach is recommended, beginning with basic Western blot analysis to confirm antibody specificity at the expected molecular weight (approximately 30.22 kDa for the core protein). Researchers should include positive controls (tissues known to express C1QTNF7, such as those indicated in the Human Protein Atlas) and negative controls (tissues or cell lines with confirmed low/no expression) .
Advanced validation methods should include immunoprecipitation followed by mass spectrometry identification, testing in cell lines with CRISPR knockout of C1QTNF7, and comparison of results across multiple antibodies targeting different epitopes of the protein. For immunohistochemistry applications, researchers should compare staining patterns with known mRNA expression data from resources like the Human Protein Atlas . Documentation of lot numbers, dilutions, incubation conditions, and complete validation protocols is essential for reproducibility.

What controls are necessary when studying C1QTNF7 in disease models?

When investigating C1QTNF7 in disease models, particularly in cancer research contexts like lung adenocarcinoma, multiple control types are essential. First, include biological controls: matched non-pathological tissues or cells from the same donor/patient when possible to account for individual genetic variation. Second, incorporate methodological controls: isotype controls for antibody-based techniques, vehicle controls for treatment studies, and empty vector controls for overexpression studies .
For studies examining C1QTNF7's role in cancer progression, time-course controls are valuable to distinguish between acute and chronic effects. When using patient-derived samples, controls should be stratified by relevant clinical factors such as age, sex, disease stage, and treatment history. If computational methods like those described in lung adenocarcinoma research are employed, technical validation using orthogonal methods is recommended, and appropriate statistical controls for multiple testing should be implemented .

How can single-cell RNA sequencing data be integrated with bulk RNA-seq to study C1QTNF7 in complex tissues?

Integration of single-cell RNA sequencing (scRNA-seq) with bulk RNA-seq provides a powerful approach to understand C1QTNF7 expression in heterogeneous tissues. Computational deconvolution techniques can help researchers estimate cellular proportions and cell-specific expression patterns. Tools like Bisque, CIBERSORT, and CIBERSORTx can be employed to decompose bulk RNA-seq data into constituent cell types, with each tool offering different advantages depending on research needs .
For optimal results, researchers should first generate high-quality reference scRNA-seq data from relevant tissues or access public datasets from repositories like DISCO, GEO, or CancerSEA. Next, apply cell-type annotation using tools like CELLiD to identify distinct cell populations. Once cell types are defined, packages like BisqueRNA (version v1.0.5) can be used to estimate cell proportions in bulk samples. This approach allows researchers to determine which cell types express C1QTNF7 and how this expression might change in disease states . For statistical robustness, researchers should consider batch effect corrections and employ cross-validation techniques to verify the accuracy of their deconvolution results.

What bioinformatic approaches can be used to investigate C1QTNF7's role in disease progression?

To investigate C1QTNF7's potential role in disease progression, particularly in contexts like cancer, several bioinformatic approaches can be employed. Survival analysis using Cox proportional hazards models can correlate C1QTNF7 expression with patient outcomes like progression-free interval (PFI). Feature selection methods such as Lasso can identify gene signatures associated with C1QTNF7 expression patterns .
Clustering algorithms (like k-means) can stratify patients based on C1QTNF7 expression in combination with other molecular features. These patient subgroups can then be analyzed for differential clinical outcomes, treatment responses, or molecular characteristics. Machine learning approaches including random forest (RF), support vector machine (SVM), extreme gradient boosting (XGBoost), and adaptive boosting (AdaBoost) can be used to build predictive models incorporating C1QTNF7 data . For validation, apply five-fold cross-validation and evaluate model performance using metrics like accuracy, recall, precision, F1 score, and area under the curve (AUC). Visualization tools like immunograms can effectively communicate complex patterns associated with C1QTNF7 expression across patient subgroups.

How can I design experiments to study the interaction between C1QTNF7 and immune cell populations?

Investigating interactions between C1QTNF7 and immune cell populations requires a multi-modal experimental approach. Begin with co-culture experiments where purified recombinant C1QTNF7 is added to different immune cell populations (T cells, macrophages, dendritic cells) isolated from peripheral blood mononuclear cells (PBMCs). Monitor changes in immune cell activation, cytokine production, and transcriptional profiles using flow cytometry, ELISA, and RNA-seq respectively .
For more complex interactions, consider using transwell systems or 3D organoid cultures that incorporate both C1QTNF7-expressing cells and immune populations. In vivo models, such as humanized immune system mice, can provide insights into systemic effects. For computational analysis, apply single-sample gene set enrichment analysis (ssGSEA) to evaluate enrichment of immune signatures in the presence of varying C1QTNF7 levels . Implement appropriate controls for each experiment, including isotype controls, vehicle treatments, and biological replicates. For mechanistic studies, consider knockdown/knockout approaches using siRNA or CRISPR to modulate C1QTNF7 expression in relevant cell types while observing consequent immune changes.

What are the optimal methods for recombinant production of C1QTNF7?

For recombinant production of C1QTNF7, E. coli expression systems have proven effective as demonstrated by commercially available recombinant proteins. The optimal expression construct typically includes the coding sequence for amino acids 17-289 of human C1QTNF7 (matching UniProtKB/Swiss-Prot entry Q9BXJ2) with an N-terminal His-tag for purification purposes . While E. coli systems are cost-effective and scalable, they produce non-glycosylated protein which may differ from native C1QTNF7 in certain functional aspects.
For applications requiring post-translational modifications, mammalian expression systems (HEK293 or CHO cells) may be more appropriate despite higher costs and lower yields. Purification should employ proprietary chromatographic techniques, typically beginning with immobilized metal affinity chromatography (IMAC) utilizing the His-tag, followed by size exclusion chromatography to remove aggregates and impurities . Protein quality should be verified through SDS-PAGE, Western blotting, and mass spectrometry. Activity validation through functional assays relevant to C1QTNF7's biological role is essential before experimental application.

How can I reliably measure C1QTNF7 expression changes in disease states?

Reliable measurement of C1QTNF7 expression changes across disease states requires a multi-level approach. At the mRNA level, quantitative RT-PCR using validated primers spanning exon junctions offers a straightforward method for relative quantification. For absolute quantification, digital PCR may provide greater precision. RNA-sequencing (bulk or single-cell) offers a broader perspective, placing C1QTNF7 expression in the context of global transcriptional changes .
At the protein level, Western blotting with validated antibodies allows semi-quantitative analysis, while ELISA or mass spectrometry-based approaches enable more precise quantification. For spatial context, immunohistochemistry or immunofluorescence can reveal expression patterns within complex tissues, though these methods require rigorous antibody validation . For longitudinal studies, consider developing a targeted proteomics assay using selected reaction monitoring (SRM) or parallel reaction monitoring (PRM). When comparing disease states, match samples for confounding variables like age, sex, and medication status, and implement appropriate normalization using stable reference genes or proteins validated specifically for the tissue and disease context under investigation.

What quasi-experimental designs are most appropriate for clinical studies involving C1QTNF7?

For clinical studies investigating C1QTNF7, several quasi-experimental designs offer robust alternatives when randomized controlled trials are impractical or unethical. Nonequivalent groups design is particularly valuable, where researchers identify patient groups with similar characteristics but differing in C1QTNF7 expression levels or mutations. This approach requires careful matching on confounding variables and statistical control of remaining differences .
Regression discontinuity designs may be applicable if clinical decisions or treatments occur based on threshold values related to C1QTNF7 or associated biomarkers, allowing comparison of patients just above and below these thresholds. Natural experiments, where external factors lead to differences in C1QTNF7 expression or function, can also provide valuable insights . For example, if a health policy change affects treatment options for conditions where C1QTNF7 is implicated, researchers could compare outcomes before and after implementation.
Time-series designs with multiple measurement points may help establish temporal relationships between C1QTNF7 expression changes and disease progression. When designing these studies, researchers should clearly acknowledge limitations in internal validity while emphasizing the higher external validity that quasi-experimental designs often offer compared to laboratory studies .

How should contradictory findings regarding C1QTNF7 function be reconciled in research?

When confronted with contradictory findings regarding C1QTNF7 function, researchers should implement a systematic approach to reconciliation. First, conduct a detailed methodological comparison, examining differences in experimental systems (cell lines, animal models, patient cohorts), protein sources (recombinant vs. native), detection methods, and statistical approaches. Document all methodological details including buffer compositions, incubation times, and equipment settings that might influence results .
Second, consider context-dependent effects where C1QTNF7 may function differently across tissue types, disease states, or in the presence of different interacting partners. Third, evaluate the role of post-translational modifications, as discrepancies may arise when comparing bacterial-expressed recombinant protein (non-glycosylated) with native forms . Meta-analysis approaches combining data across multiple studies can help identify consistent patterns despite methodological variations.
For truly irreconcilable findings, design decisive experiments specifically addressing the contradictions, ideally incorporating multiple methodologies within a single study to directly compare outcomes under standardized conditions. Throughout this process, maintain scientific objectivity and avoid confirmation bias by being equally critical of results supporting and contradicting your hypothesis.

What statistical approaches are recommended for analyzing C1QTNF7 expression in heterogeneous tissue samples?

When analyzing C1QTNF7 expression in heterogeneous tissue samples, statistical approaches must account for cellular composition differences that may confound results. Computational deconvolution methods should be employed to estimate cellular proportions before expression analysis. Tools like BisqueRNA can integrate single-cell reference data with bulk RNA-seq to provide cell-type-specific expression estimates .
For differential expression analysis, linear mixed-effects models can account for both fixed effects (disease status, treatment) and random effects (patient characteristics, batch). When cellular proportions are available, include these as covariates in statistical models to distinguish between true expression changes and shifts in cellular composition. For identifying co-expression patterns, weighted gene co-expression network analysis (WGCNA) can reveal modules of genes with similar expression patterns across samples .
Machine learning approaches like random forest or support vector machines can identify complex patterns associated with C1QTNF7 expression. Cross-validation (e.g., five-fold) should be employed to assess model robustness, and performance metrics including accuracy, recall, precision, F1 score, and area under the curve (AUC) should be reported comprehensively . For all analyses, implement appropriate multiple testing corrections (e.g., Benjamini-Hochberg procedure) to control false discovery rates.

How can researchers determine if C1QTNF7 has potential as a biomarker in disease studies?

Evaluating C1QTNF7's potential as a disease biomarker requires a structured, multi-phase assessment approach. Initial discovery phase should identify significant associations between C1QTNF7 expression/levels and disease states using retrospective samples, applying methods like Cox proportional hazards models to correlate with clinical outcomes such as progression-free interval (PFI) . Adjust for established risk factors and potential confounders in these analyses.
Validation phase should test predetermined C1QTNF7 thresholds in independent cohorts, calculating sensitivity, specificity, positive predictive value, negative predictive value, and area under ROC curve. Compare performance against existing biomarkers using net reclassification improvement (NRI) and integrated discrimination improvement (IDI) metrics. For clinical utility assessment, determine if C1QTNF7 measurements would alter clinical decisions by calculating decision curve analysis metrics . Analytical validation must establish assay precision, accuracy, reportable range, and pre-analytical requirements (sample collection, processing, storage conditions). Technological assessment should compare measurement platforms (ELISA, mass spectrometry, qPCR) for cost-effectiveness and practicality in clinical settings. Throughout this process, adhere to STARD (Standards for Reporting of Diagnostic Accuracy) guidelines and consider regulatory requirements for clinical biomarker implementation.

Product Science Overview

Introduction

Complement C1q Tumor Necrosis Factor-Related Protein 7 (CTRP7) is a member of the C1q/TNF-related protein (CTRP) family. This family consists of secreted proteins that share structural similarities with the complement component C1q and the tumor necrosis factor (TNF) superfamily. CTRPs are known for their roles in various physiological processes, including metabolism, inflammation, and immune responses .

Structure

CTRP7, like other members of the CTRP family, is composed of several distinct domains:

  1. N-terminal signal peptide: This sequence directs the protein to the secretory pathway.
  2. Variable domain: This region varies among different CTRP family members 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 responsible for the trimerization of the protein.
  4. C-terminal globular C1q (gC1q) domain: This domain is homologous to the globular domain of the complement protein C1q and is crucial for the protein’s biological activity .
Function

CTRP7 has been implicated in several biological processes:

  • Metabolism: CTRP7 is involved in the regulation of lipid and glucose metabolism. It enhances insulin sensitivity and has anti-inflammatory properties, making it a potential therapeutic target for metabolic disorders such as obesity and type 2 diabetes .
  • Inflammation: CTRP7 plays a role in modulating inflammatory responses. It can influence the production of pro-inflammatory cytokines and has been shown to have protective effects against inflammatory diseases .
  • Immune Response: The protein is also involved in the immune response, contributing to the regulation of immune cell functions and the maintenance of immune homeostasis .
Recombinant Production

Human recombinant CTRP7 is produced using recombinant DNA technology. This involves the insertion of the gene encoding CTRP7 into an expression vector, which is then introduced into a host cell (such as E. coli or mammalian cells). The host cells express the protein, which is subsequently purified for research or therapeutic use. Recombinant production allows for the generation of large quantities of the protein with high purity and consistency .

Clinical Implications

Given its roles in metabolism and inflammation, CTRP7 is being studied for its potential therapeutic applications. It may serve as a biomarker for metabolic and inflammatory diseases and could be targeted for the development of new treatments. Research is ongoing to better understand the mechanisms by which CTRP7 exerts its effects and to explore its potential in clinical settings .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.