RBP7 Human

Retinol Binding Protein-7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Adipogenesis and Retinol Metabolism

  • Adipocyte Differentiation: RBP7 overexpression in 3T3-L1 preadipocytes enhances triglyceride accumulation and upregulates adipogenic markers (Pparγ, Fabp4, C/ebpα) . Knockdown reduces differentiation, rescued by retinoic acid (RA) .

  • RA Homeostasis: RBP7 modulates RA synthesis (Raldh1↑) and catabolism (Cyp26a1↑), balancing nuclear RA levels via feedback loops .

  • Cold Adaptation: RBP7 expression increases in brown adipose tissue during cold exposure, linking it to thermogenesis .

Endothelial Function

  • PPARγ Cofactor: RBP7 is an endothelial-specific PPARγ target gene. It mediates PPARγ’s antioxidant effects by upregulating adiponectin, reducing oxidative stress .

  • Vascular Protection: RBP7-deficient mice develop endothelial dysfunction under high-fat diets or angiotensin II stress, reversed by superoxide scavengers .

Cancer Prognosis

  • Colon Cancer: High RBP7 expression correlates with poor survival (HR = 2.54, p = 0.009 in early-stage; HR = 2.5, p = 0.038 in TCGA data) .

  • Mechanistic Link: RBP7 associates with epithelial-mesenchymal transition (EMT) markers (ZEB1, ZEB2↑; CDH1↓) and invasiveness .

Biomarker Potential

Clinical ParameterAssociation with RBP7Study
Tumor StageHigher T-category (T3/T4 vs. T1/T2) and nodal metastasis (p < 0.001)
SurvivalIndependent predictor of poor cancer-specific survival (HR = 2.5–2.54)
Therapeutic TargetPotential for obesity and metabolic syndrome via PPARγ/RARE pathway modulation

Recombinant RBP7 in Research

  • Applications: Used in SDS-PAGE, mass spectrometry, and retinol transport assays .

  • Limitations: Research-use only; not for diagnostic/therapeutic use .

Key Research Findings

  • Adipogenesis: RBP7 knockout mice show impaired adipocyte differentiation, reversed by RA .

  • Endothelial Dysfunction: RBP7 deficiency exacerbates oxidative stress in vasculature under metabolic stress .

  • Cancer Progression: RBP7-high colon tumors exhibit aggressive phenotypes and resistance to therapy .

Future Directions

  • Mechanistic Studies: Elucidate RBP7’s role in RA nuclear transport and PPARγ crosstalk.

  • Therapeutic Development: Target RBP7 in obesity or colon cancer via small-molecule inhibitors or gene editing.

  • Clinical Trials: Validate RBP7 as a prognostic biomarker in multicenter cohorts.

Product Specs

Introduction
RBP7, a member of a superfamily of small cytoplasmic proteins, interacts with hydrophobic ligands. This cytoplasmic protein, similar to CRBP I and CRBP II, forms β-barrel structures and plays a role in the intracellular transport of retinol. RBP7, a recently identified cellular retinol carrier, is found in the kidney, heart, and transverse colon in humans.
Description
Recombinant human RBP7, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It contains 154 amino acids (including a 20 amino acid His tag at the N-terminus, spanning residues 1-134) and has a molecular weight of 17.6 kDa. Purification of RBP7 is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The RBP7 solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 2mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability during frozen storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
Retinoid-binding protein 7, Cellular retinoic acid-binding protein 4, CRABP4, CRBP4, Cellular retinoic acid-binding protein IV, CRABP-IV, RBP7, MGC70641.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MPADLSGTWT LLSSDNFEGY MLALGIDFAT RKIAKLLKPQ KVIEQNGDSF TIHTNSSLRN YFVKFKVGEE FDEDNRGLDN RKCKSLVIWD NDRLTCIQKG EKKNRGWTHW IEGDKLHLEM FCEGQVCKQT FQRA.

Q&A

What is RBP7 and what are its primary functions in human cells?

RBP7 (Retinol-binding protein 7) is a member of the cellular retinol-binding protein (CRBP) family involved in vitamin A uptake, storage, and metabolism . It functions primarily in the transport and metabolism of retinol (vitamin A), playing crucial roles in cell differentiation pathways . RBP7 belongs to the family of intracellular lipid- and fatty acid-binding proteins (FABPs) . Its activity is particularly important in retinoid signaling, where it facilitates the availability of retinoic acid for gene regulation and helps maintain retinol homeostasis .

In which human tissues is RBP7 predominantly expressed?

RBP7 demonstrates highly tissue-specific expression patterns. Recent evidence suggests that RBP7 is an endothelium-enriched transcript, with some data indicating it may be exclusively expressed in endothelium, particularly in small blood vessels . In mice, Rbp7 is abundantly expressed in both white and brown adipose tissues, with higher expression in adipocytes compared to the stromal vascular fraction . This tissue-specific pattern makes RBP7 particularly interesting as a potential endothelium-specific biomarker and therapeutic target .

How is RBP7 regulated at the transcriptional level?

RBP7 is a direct target gene of peroxisome proliferator-activated receptor-γ (PPARγ), a master regulator of lipid metabolism and adipogenesis . Experimental evidence shows that RBP7 mRNA expression is induced by PPARγ agonists like rosiglitazone, and this induction can be blunted by pretreatment with PPARγ antagonists such as GW9662 . Additionally, RBP7 protein levels in mouse lung endothelial cells (MLECs) increase in response to either rosiglitazone treatment or PPARγ overexpression, with synergistic effects when both conditions are present .

What is the prognostic value of RBP7 in cancer research?

RBP7 has significant prognostic value in several cancer types. In breast cancer, particularly ER-positive subtypes, lower expression of RBP7 correlates with worse prognosis . Conversely, in colon cancer, high expression of RBP7 serves as an independent biomarker of poor cancer-specific survival in both early and late-stage disease . These contrasting roles highlight the context-dependent nature of RBP7's function in different cancer types and underscore the importance of tissue-specific analysis when evaluating RBP7 as a prognostic marker.

How does RBP7 contribute to breast cancer progression?

In breast cancer, RBP7 expression is frequently downregulated through promoter hypermethylation . This methylation-mediated silencing appears to promote breast cancer progression, particularly in ER-positive subtypes. Functional enrichment analysis demonstrates that downregulation of RBP7 expression may exert its biological influence on breast cancer through the PPAR pathway and the PI3K/AKT pathway . These pathways are critically involved in cell proliferation, survival, and metabolism, explaining the mechanism by which RBP7 silencing contributes to cancer progression.

What is the relationship between RBP7 and colon cancer invasion?

High expression of RBP7 in colon cancer is linked to tumor progression and poor survival outcomes . Gene set enrichment analysis has revealed a strong association between RBP7, colon cancer invasion, and epithelial-mesenchymal transition (EMT) . Functional studies demonstrate that ectopic expression of RBP7 increases migration and invasion of colon cancer cells, suggesting that RBP7 may actively promote metastatic potential in this cancer type . This finding contrasts with RBP7's apparent tumor-suppressive role in breast cancer, highlighting the complex, context-dependent functions of this protein in different cancer types.

How does RBP7 influence adipogenesis and lipid metabolism?

RBP7 plays a critical role in promoting adipogenesis. In experimental models, Rbp7 overexpression promotes 3T3-L1 preadipocyte differentiation, resulting in increased triglyceride accumulation and upregulation of adipogenic markers including Pparγ, Fabp4, C/ebpα, and AdipoQ . Conversely, Rbp7-deficient adipocytes show impaired differentiation, an effect that can be rescued by retinoic acid (RA) supplementation . These findings indicate that RBP7 mediates its effects on adipogenesis at least partly through regulation of retinoid signaling and availability.

What is the relationship between RBP7 and retinoic acid signaling in adipocytes?

RBP7 significantly impacts retinoic acid (RA) signaling in adipocytes. Indirect assessment using RAR response element (RARE)-Luc reporter assays demonstrates that Rbp7 overexpression significantly increases RARE-Luc reporter activity, indicating enhanced RA-dependent transcriptional activation . Additionally, Rbp7 overexpression alters the expression of genes involved in retinol metabolism, including increased expression of Raldh1 (responsible for RA production) and upregulation of Lrat and Cyp26a1 (involved in retinol storage and RA catabolism, respectively) . This suggests that RBP7 increases nuclear RA levels and regulates gene expression related to retinol homeostasis.

What experimental approaches are most effective for studying RBP7's role in adipogenesis?

The most effective experimental approaches for studying RBP7's role in adipogenesis combine genetic manipulation with functional readouts. Key methodologies include:

  • Gene expression modulation: Using viral vectors for overexpression or siRNA for knockdown of Rbp7 in preadipocyte cell lines such as 3T3-L1 .

  • Differentiation assays: Monitoring triglyceride accumulation and expression of adipogenic markers (Pparγ, Fabp4, C/ebpα, AdipoQ) following differentiation induction .

  • Rescue experiments: Supplementing Rbp7-deficient cells with retinoic acid to determine whether the effects are mediated through retinoid signaling .

  • Reporter assays: Using RARE-Luc reporters to assess nuclear RA levels and transcriptional activity .

  • qPCR and Western blot analysis: Quantifying changes in expression of genes involved in retinoid metabolism and adipogenesis .

These approaches allow for comprehensive analysis of both the phenotypic consequences of RBP7 modulation and the underlying molecular mechanisms.

What is the role of RBP7 in endothelial function?

RBP7 serves as an endothelium-specific PPARγ cofactor that is essential for vascular protection under stress conditions . While RBP7-deficient mice exhibit normal endothelial function at baseline, they develop severe endothelial dysfunction when exposed to cardiovascular stressors, such as high-fat diet and subpressor angiotensin II . This dysfunction occurs through an oxidative stress-dependent mechanism and can be rescued by superoxide scavengers . These findings identify RBP7 as a critical mediator of PPARγ's protective effects in the endothelium, particularly under conditions of metabolic or cardiovascular stress.

How does RBP7 interact with PPARγ in the endothelium?

RBP7 functions as an endothelium-specific PPARγ cofactor, mediating the induction of a subset of PPARγ target genes in response to PPARγ agonists like rosiglitazone . RNA sequencing revealed that RBP7 is required for the induction of certain PPARγ target genes in the endothelium, including adiponectin . This suggests that RBP7 may play a selective role in facilitating PPARγ-dependent transcription, potentially by modulating the availability of retinoids that can influence nuclear receptor activity. The endothelium-specific expression of RBP7 makes it a particularly important mediator of PPARγ's vascular protective effects.

What experimental models are appropriate for investigating RBP7's vascular functions?

Based on the research literature, the following experimental models are most appropriate for investigating RBP7's vascular functions:

  • Genetic mouse models: RBP7-deficient mice provide a valuable tool for studying endothelial cell-specific RBP7 function in vivo, particularly when challenged with stressors like high-fat diet or angiotensin II .

  • Ex vivo vessel reactivity: Assessing endothelium-dependent relaxation in isolated vessels (e.g., basilar artery) in response to acetylcholine provides functional readouts of endothelial health .

  • Cultured endothelial cells: Immortalized mouse lung endothelial cells (MLECs) that endogenously express RBP7 allow for in vitro manipulation and detailed mechanistic studies .

  • Stress models: High-fat diet feeding and angiotensin II infusion serve as physiologically relevant stressors that reveal RBP7's role in protecting against endothelial dysfunction .

  • Pharmacological interventions: Using PPARγ agonists (rosiglitazone) and antagonists (GW9662) helps elucidate the relationship between PPARγ signaling and RBP7 function .

These models collectively provide a comprehensive toolkit for examining RBP7's role in endothelial biology across different experimental contexts.

How can methylation of the RBP7 promoter be effectively analyzed?

Analysis of RBP7 promoter methylation requires a multi-faceted approach:

  • Bisulfite sequencing: The gold standard for methylation analysis, allowing single-nucleotide resolution of methylation patterns in the RBP7 promoter region.

  • Methylation-specific PCR (MSP): A targeted approach to rapidly assess methylation status at specific CpG sites within the RBP7 promoter.

  • Pyrosequencing: Providing quantitative methylation analysis with the ability to assess multiple CpG sites simultaneously.

  • Genome-wide methylation arrays: Useful for comparing RBP7 promoter methylation across different sample types or in response to treatments.

  • Integration with expression data: Correlating methylation patterns with RBP7 mRNA and protein expression levels to establish functional relationships .

In breast cancer research specifically, methylation analysis should focus on the RBP7 promoter region, as hypermethylation correlates with gene silencing and poorer prognosis in ER-positive patients .

What approaches should be used to study RBP7's role in cancer invasion and EMT?

Studying RBP7's role in cancer invasion and EMT requires a comprehensive approach combining multiple methodologies:

  • Invasion and migration assays: Transwell assays are effective for quantifying the impact of RBP7 modulation on cancer cell invasiveness .

  • Gene expression profiling: RNA sequencing or microarray analysis to identify EMT-related genes affected by RBP7 expression changes.

  • EMT marker analysis: Western blotting or immunofluorescence to assess expression of epithelial markers (E-cadherin, ZO-1) and mesenchymal markers (N-cadherin, vimentin) following RBP7 manipulation.

  • Gene set enrichment analysis (GSEA): To identify pathways and biological processes associated with RBP7 expression in cancer contexts .

  • In vivo metastasis models: Xenograft studies tracking metastatic spread in animals with RBP7-modified cancer cells.

These approaches collectively provide mechanistic insights into how RBP7 influences cancer progression, particularly in colon cancer where high RBP7 expression is associated with invasion and poor prognosis .

How can researchers effectively identify and validate RBP7 target genes?

Identifying and validating RBP7 target genes requires a multi-layered approach:

  • Chromatin immunoprecipitation followed by sequencing (ChIP-seq): While RBP7 itself is not a transcription factor, ChIP-seq for PPARγ or other transcription factors in RBP7-modulated systems can reveal altered binding patterns.

  • RNA sequencing: Comparing transcriptomes between RBP7-overexpressing, knockdown, and control conditions identifies differentially expressed genes .

  • Pathway analysis: Using tools like KEGG or Reactome to identify pathways enriched among RBP7-responsive genes .

  • qPCR validation: Confirming expression changes of key candidates identified through global approaches.

  • Reporter assays: Using constructs like RARE-Luc to assess functional impacts on specific pathways .

  • Rescue experiments: Determining whether reintroduction of candidate genes can rescue phenotypes observed in RBP7-deficient models.

These approaches have successfully identified RBP7-dependent gene expression programs in contexts such as adipogenesis and endothelial function .

How should contradictory findings about RBP7's role in different cancer types be resolved?

The seemingly contradictory findings regarding RBP7's role in different cancer types—tumor suppressive in breast cancer but oncogenic in colon cancer—should be approached through:

  • Tissue-specific context analysis: Recognizing that RBP7 functions within different cellular environments and signaling networks in different tissues.

  • Molecular subtyping: Further stratifying analyses within cancer types (e.g., ER-positive versus ER-negative breast cancer) .

  • Multi-omics integration: Combining data on expression, methylation, mutation, and pathway activation to build comprehensive models of RBP7 function.

  • Consideration of retinoid signaling differences: Evaluating how retinoid metabolism and signaling differ between tissue types.

  • Mechanistic studies in multiple cancer models: Performing parallel experiments in breast and colon cancer models to directly compare RBP7's effects on common endpoints.

These approaches acknowledge that RBP7's function is likely context-dependent, influenced by tissue-specific factors, disease stage, and molecular subtype .

What statistical approaches are most appropriate for correlating RBP7 expression with clinical outcomes?

The most appropriate statistical approaches for correlating RBP7 expression with clinical outcomes include:

These methods have been successfully applied in studies examining RBP7's prognostic significance in breast and colon cancer, where expression levels correlate with distinct survival outcomes .

How can researchers distinguish direct effects of RBP7 from indirect consequences in pathway analyses?

Distinguishing direct effects of RBP7 from indirect consequences requires:

  • Temporal analysis: Examining early versus late gene expression changes following RBP7 modulation.

  • Dose-response relationships: Testing whether gene expression changes correlate with the degree of RBP7 modulation.

  • Rescue experiments: Determining whether specific pathway alterations can be reversed by restoring RBP7 levels or function.

  • Biochemical interaction studies: Identifying proteins that directly interact with RBP7 through co-immunoprecipitation or proximity labeling.

  • In silico pathway modeling: Constructing mathematical models of RBP7-affected pathways to predict direct versus indirect relationships.

These approaches help establish causality and elucidate the mechanisms by which RBP7 influences pathways such as PPAR and PI3K/AKT in cancer contexts or retinoid signaling in adipocytes .

How might targeting RBP7 be exploited for potential therapeutic interventions?

Targeting RBP7 for therapeutic purposes could take several approaches:

  • Cancer treatment stratification: Using RBP7 expression or methylation status to guide treatment decisions, particularly in ER-positive breast cancer where low RBP7 correlates with poor prognosis .

  • Demethylating agents: Exploring drugs that reverse DNA methylation to restore RBP7 expression in breast cancer contexts where it is silenced .

  • RBP7 inhibitors: Developing small molecules that disrupt RBP7 function could be beneficial in colon cancer, where high RBP7 expression promotes invasion .

  • Metabolic disease applications: Given RBP7's role in adipogenesis, targeting its function could provide a novel approach for obesity therapy .

  • Vascular protection: Enhancing RBP7 function in endothelial cells could protect against stress-induced endothelial dysfunction in cardiovascular disease contexts .

These potential interventions highlight the context-dependent nature of RBP7 targeting, with opposing strategies potentially required for different disease states.

What are the challenges in developing RBP7-targeted therapies?

Developing RBP7-targeted therapies faces several significant challenges:

  • Tissue specificity: RBP7's predominant expression in endothelium and its diverse functions in different tissues necessitate highly targeted delivery approaches .

  • Context-dependent function: RBP7's apparently opposing roles in breast versus colon cancer require careful consideration of disease context .

  • Involvement in fundamental processes: RBP7's role in retinoid metabolism could lead to unintended consequences if broadly inhibited .

  • Limited structural information: Incomplete understanding of RBP7's three-dimensional structure and binding mechanisms hampers rational drug design.

  • Biomarker validation: More extensive validation of RBP7 as a prognostic or predictive biomarker is needed before clinical implementation.

Addressing these challenges requires continued basic research into RBP7's structure, function, and tissue-specific roles, alongside development of targeted delivery systems to minimize off-target effects.

How can RBP7 expression be effectively restored in contexts where it is silenced?

In contexts like breast cancer where RBP7 is silenced through promoter hypermethylation, several approaches could potentially restore expression:

  • DNA methyltransferase inhibitors: Drugs like 5-azacytidine or decitabine that inhibit DNA methylation could reverse RBP7 promoter hypermethylation .

  • PPARγ agonists: Since RBP7 is a PPARγ target gene, treatment with PPARγ agonists like rosiglitazone might increase RBP7 expression .

  • Histone deacetylase inhibitors: These epigenetic modulators could potentially enhance RBP7 expression by promoting an open chromatin state.

  • Gene therapy approaches: Viral vector-mediated delivery of RBP7 could restore expression in cells where it is silenced.

  • CRISPR-based epigenome editing: Targeted demethylation of the RBP7 promoter using CRISPR-dCas9 systems coupled with TET enzymes.

These strategies could be particularly relevant for ER-positive breast cancer patients, where RBP7 silencing correlates with worse prognosis and might contribute to disease progression .

What are the most promising avenues for future RBP7 research?

The most promising future research directions for RBP7 include:

  • Structural biology: Determining the three-dimensional structure of RBP7 and how it interacts with retinoids and protein partners.

  • Single-cell analyses: Investigating cell-type specific functions of RBP7, particularly within heterogeneous tissues like tumors.

  • In vivo models: Generating tissue-specific RBP7 knockout or overexpression models to dissect its role in different physiological contexts.

  • Clinical biomarker validation: Large-scale studies validating RBP7's utility as a prognostic or predictive biomarker in various cancer types .

  • Therapeutic targeting: Developing and testing compounds that modulate RBP7 function or expression in disease-relevant contexts.

These research directions would significantly advance our understanding of RBP7's biological functions and therapeutic potential across multiple disease contexts.

How might advances in single-cell technologies enhance our understanding of RBP7 function?

Single-cell technologies offer transformative potential for understanding RBP7 function through:

  • Cell-type resolution: Precisely defining which endothelial subtypes express RBP7 and how expression varies across vascular beds .

  • Heterogeneity analysis: Examining whether RBP7 expression is uniform or heterogeneous within tissues, particularly in cancer contexts.

  • Trajectory analysis: Tracking how RBP7 expression changes during processes like adipogenesis or endothelial dysfunction .

  • Multi-omics integration: Combining single-cell transcriptomics, epigenomics, and proteomics to build comprehensive models of RBP7 regulation and function.

  • Spatial context: Using spatial transcriptomics to understand how RBP7-expressing cells interact with their microenvironment.

These approaches would provide unprecedented insights into RBP7's context-specific functions and potentially reveal novel therapeutic opportunities.

What interdisciplinary approaches might yield new insights into RBP7 biology?

Novel insights into RBP7 biology could emerge from interdisciplinary approaches including:

  • Systems biology: Integrating multiple data types to model RBP7's role in complex networks like retinoid metabolism and PPARγ signaling .

  • Chemical biology: Developing small-molecule probes to manipulate RBP7 function with temporal and spatial precision.

  • Computational biology: Using machine learning to identify patterns in RBP7-associated gene expression across different contexts.

  • Metabolomics: Profiling retinoid metabolites in RBP7-modulated systems to understand its impact on vitamin A processing.

  • Translational research: Bridging basic RBP7 biology with clinical applications in cancer diagnostics and therapeutics .

Product Science Overview

Gene and Protein Structure

The RBP7 gene is located on human chromosome 1p36.22 and encodes a protein consisting of 134 amino acids . The protein binds all-trans-retinol, although it has a lower binding affinity compared to other CRBPs . This binding is vital for the intracellular transport of retinol, ensuring its availability for metabolic processes .

Function and Importance

RBP7 plays a significant role in several biological processes, including:

  • Vitamin A Metabolism: It helps in the transport and stability of retinol, which is essential for vision, immune function, and cellular growth .
  • Adipogenesis: RBP7 is involved in the formation of adipose (fat) tissue .
  • Response to Cold Exposure: It plays a role in the body’s response to cold environments .
  • Nutritional Treatment: RBP7 is important in the body’s adaptation to different nutritional states .
Clinical Relevance

RBP7 has been studied for its potential role in various health conditions:

  • Hypertension: It has been implicated in the regulation of blood pressure .
  • Cancer: RBP7 functions as a tumor suppressor in hormone receptor-positive breast cancer .
  • Endothelial Function: It is an endothelium-specific gene regulated by PPARγ, a nuclear receptor involved in fat metabolism and inflammation .
Research and Applications

Human recombinant RBP7 is used in research to study its function and potential therapeutic applications. By understanding how RBP7 interacts with retinol and other molecules, scientists can develop new treatments for diseases related to vitamin A deficiency, metabolic disorders, and certain types of cancer .

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.