RBP1 Human

Retinol Binding Protein-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Retinol Transport and Homeostasis

RBP1 facilitates retinol uptake by accepting it from the transport protein STRA6, ensuring retinol delivery to peripheral tissues for metabolic activation into retinoic acid . This process is critical for:

  • Epithelial differentiation

  • Embryonic development

  • Vision cycle regulation

Nuclear Signaling Modulation

Beyond transport, RBP1 acts as a chaperone to regulate retinoic acid availability for nuclear receptors (RARs/RXRs), influencing gene expression linked to cell differentiation and apoptosis .

Oncogenic Roles

  • Oral Squamous Cell Carcinoma (OSCC): RBP1 overexpression correlates with advanced TNM stages, lymphatic metastasis, and poor prognosis. In vitro, RBP1 enhances SCC15 cell proliferation (2.1-fold increase in colony formation) and invasion via autophagy activation through CKAP4 interaction .

  • Endometrial Cancer: Loss of RBP1 expression disrupts retinol bioavailability, promoting malignant transformation .

Cardiovascular and Immune Links

  • Coronary Artery Disease (CAD): RBP1 hypermethylation in atherosclerosis patients correlates with immune cell infiltration patterns (e.g., CD4+ T cells and macrophages) .

  • Pan-Cancer Analysis: RBP1 expression associates with stromal/immune scores in prostate adenocarcinoma (PRAD) and ovarian cancer (OV), suggesting immune microenvironment modulation .

Table 2: Key Research Findings on RBP1

Study FocusKey OutcomeSource
OSCC PathogenesisRBP1 knockout reduces xenograft tumor volume by 58%
CAD Immune ResponseRBP1 positively correlates with CD4+ T cells (ρ=0.42, p<0.01)
Retinol HomeostasisCRBP1 deficiency increases retinol oxidation by 3.7-fold

Research Applications

Recombinant human RBP1 proteins are widely used in biochemical assays. Key commercial variants include:

Product Specs

Introduction
RBP1, a member of the calycin superfamily and fatty-acid binding protein (FABP) family, is the carrier protein responsible for transporting retinol (vitamin A alcohol) from the liver to peripheral tissues. It also acts as a bridging molecule, recruiting histone deacetylases (HDACs), which are crucial for gene expression regulation. RBP1 is present in most tissues, with higher concentrations found in the pancreas, adrenal gland, pituitary gland, fetal liver, and adult ovary.
Description
Recombinant human RBP1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 220 amino acids (1-197 a.a.). With a molecular weight of 24.7kDa, it includes a 23 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Colorless, sterile filtered solution.
Formulation
The RBP1 protein solution (0.5mg/ml) is supplied in 20mM Tris-HCl buffer (pH 8.0), containing 200mM NaCl, 2mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Retinol binding protein 1 cellular, CRBP, CRBP1, CRABP-I, RBPC.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMDPPAGF VRAGNPAVAA PQSPLSPEGA HFRAAHHPRS TGSRCPGSLQ PSRPLVANWL QSLPEMPVDF TGYWKMLVNE NFEEYLRALD VNVALRKIAN LLKPDKEIVQ DGDHMIIRTL STFRNYIMDF QVGKEFEEDL TGIDDRKCMT TVSWDGDKLQ CVQKGEKEGR GWTQWIEGDE LHLEMRVEGV VCKQVFKKVQ

Q&A

What is the molecular structure of human RBP1?

Human RBP1 is a relatively small intracellular protein with a molecular weight of approximately 16 kDa, encoded by the RBP1 gene located on chromosome 3. The gene harbors four exons encoding 24, 59, 33, and 16 amino acid residues respectively, with the second intervening sequence alone occupying 19 kb of the 21 kb gene . RBP1 belongs to the family of intracellular lipid-binding proteins and has a characteristic β-barrel structure with a central binding pocket that accommodates retinol. This specific three-dimensional configuration enables RBP1 to securely bind retinol while navigating through the aqueous cellular environment. When studying RBP1 structure, researchers should employ techniques such as crystallography or NMR spectroscopy to understand the precise binding interactions with retinol and potential protein partners.

What are the primary functions of RBP1 in human cellular physiology?

RBP1 serves as a cytosolic carrier protein primarily responsible for regulating retinol homeostasis in human cells. Its main functions include: (1) accepting retinol from the transmembrane transport protein STRA6, facilitating cellular uptake of retinol ; (2) intracellular transport of retinol to specific cellular compartments where it is metabolized or stored; (3) contributing to retinoid homeostasis by regulating the bioavailability of retinol for conversion to retinoic acid; (4) protecting bound retinol from degradation in the aqueous cytosolic environment; and (5) participating in the maintenance of the differentiative state of certain cell types, including endometrial cells . Research approaches to study these functions should include fluorescently-tagged retinol tracking in cells with normal versus altered RBP1 expression, and metabolic tracing experiments using isotope-labeled retinol to quantify the impact of RBP1 on retinol metabolism.

How is RBP1 expression regulated in different tissue types?

RBP1 expression varies significantly across human tissues due to complex regulatory mechanisms. These include: (1) tissue-specific transcription factors that bind to the RBP1 promoter region; (2) epigenetic modifications such as DNA methylation and histone modifications that alter chromatin accessibility; (3) post-transcriptional regulation through microRNAs; (4) feedback regulation by retinoid levels themselves; and (5) developmental programming that establishes tissue-specific expression patterns. When analyzing RBP1 expression patterns, researchers should employ multiple methodologies including RNA-seq, qRT-PCR, western blotting, and immunohistochemistry to obtain comprehensive data. Tissue microarrays with antibodies such as ab154881 (effective at 1/100 dilution for IHC) can provide spatial information about RBP1 distribution. Comparative studies should always include appropriate normalization controls and multiple reference tissues.

What cellular processes depend on proper RBP1 function?

Several critical cellular processes depend on proper RBP1 function: (1) vitamin A metabolism, including the conversion of retinol to retinal and subsequently to retinoic acid; (2) cellular differentiation, particularly in epithelial tissues where retinoic acid signaling is crucial; (3) maintenance of the differentiated phenotype in tissues such as the endometrium, where loss of RBP1 is associated with cancer development ; (4) possibly autophagy regulation, as evidenced by the RBP1-CKAP4 axis in OSCC ; and (5) protection against oxidative stress through regulation of retinoid antioxidant functions. To study the impact of RBP1 on these processes, researchers should employ gene silencing approaches (siRNA, shRNA, or CRISPR-Cas9) coupled with comprehensive phenotypic and molecular analyses. Multiparametric assays that simultaneously measure differentiation markers, autophagy flux, and cell survival will provide the most informative results.

How does RBP1 interact with other proteins in the retinoid signaling pathway?

RBP1 engages in several key protein-protein interactions within the retinoid signaling pathway: (1) with STRA6, the membrane receptor that facilitates retinol uptake, where RBP1 acts as the intracellular acceptor ; (2) with retinol-metabolizing enzymes that convert retinol to retinal; (3) with CKAP4 (cytoskeleton-associated protein 4), forming the RBP1-CKAP4 axis that regulates autophagy in cancer cells ; (4) potentially with nuclear retinoid receptors or their cofactors, influencing retinoid-dependent gene expression; and (5) with cytoskeletal components that may facilitate intracellular trafficking. To study these interactions, researchers should employ co-immunoprecipitation with antibodies like ab154881 , proximity ligation assays to visualize interactions in situ, and fluorescence resonance energy transfer (FRET) to monitor dynamic interactions in living cells. Yeast two-hybrid screening can identify novel RBP1 interaction partners.

How is RBP1 expression altered in different cancer types?

RBP1 expression shows remarkable context-dependent alterations across cancer types: (1) significant upregulation in oral squamous cell carcinoma (OSCC), where it was identified as one of the most significantly upregulated differentially expressed proteins (DEPs) with >2-fold change compared to normal tissues ; (2) downregulation in endometrial cancer, where loss of CRBP-1 (RBP1) is associated with cancer development ; (3) variable expression patterns in other cancer types, suggesting tissue-specific roles; (4) correlation with malignant phenotypes in OSCC, including poor differentiation, advanced TNM stage, and lymphatic metastasis ; and (5) altered subcellular localization in some cancer cells. When investigating RBP1 in cancer, researchers should employ matched tumor-normal pairs from the same patients, utilize multiple detection methods (qRT-PCR, western blot, immunohistochemistry), and correlate expression with comprehensive clinicopathological data. These context-dependent expression patterns suggest RBP1 may function as either a tumor suppressor or oncogene depending on cellular context.

What is the relationship between RBP1 and oral squamous cell carcinoma (OSCC)?

The relationship between RBP1 and OSCC is multifaceted: (1) RBP1 is significantly upregulated in OSCC tissues and cell lines compared to normal controls, as demonstrated by iTRAQ-based proteomics analysis coupled with 2D LC-MS/MS ; (2) RBP1 overexpression promotes cancer cell growth, migration, and invasion in SCC15 cells in vitro ; (3) silencing RBP1 suppresses tumor formation in xenografted mice ; (4) RBP1 expression levels positively correlate with malignant phenotypes including differentiation status, TNM stage, and lymphatic metastasis ; and (5) mechanistically, RBP1 promotes oncogenesis through activation of the RBP1-CKAP4 axis, which regulates autophagy . For OSCC research, SCC15, SCC25, SCC9, and CAL27 cell lines have been validated as appropriate models, with SCC15 showing particularly high RBP1 expression. Silencing RBP1 with siRNA technology has been effective in demonstrating its oncogenic properties in these models.

How does the RBP1-CKAP4 axis activate oncogenic autophagy?

The RBP1-CKAP4 axis represents a critical regulatory mechanism for autophagy in cancer cells: (1) RBP1 interacts with CKAP4 (cytoskeleton-associated protein 4), forming a functional complex ; (2) this interaction triggers the activation of autophagic machinery in OSCC cells, promoting cancer cell survival ; (3) RBP1-mediated autophagy confers advantages for cancer cell growth, migration, and invasion ; (4) experimental inactivation of autophagy rescues the RBP1-CKAP4-mediated malignant biological behaviors of OSCC cells ; and (5) this axis provides a mechanistic link between primary oncogenic features and autophagy induction. To study this pathway, researchers should employ autophagy flux assays (measuring LC3-II/LC3-I ratio and p62 degradation), co-immunoprecipitation to confirm the physical interaction, and genetic approaches to manipulate both proteins individually and in combination. Confocal microscopy with fluorescently-tagged proteins can visualize the spatial relationship between RBP1, CKAP4, and autophagosomal structures.

What is the contrasting role of RBP1 in endometrial cancer?

RBP1 exhibits a contrasting role in endometrial cancer compared to OSCC: (1) in normal endometrium, RBP1 contributes to maintaining the differentiative state of endometrial cells through regulation of retinol bioavailability ; (2) unlike in OSCC, loss of CRBP-1 (RBP1) expression is associated with endometrial cancer development ; (3) this suggests a potential tumor suppressor role in the endometrial context, contrasting with its oncogenic properties in OSCC; (4) the loss of RBP1 may compromise retinoid signaling, which normally promotes differentiation and inhibits abnormal proliferation; and (5) these contrasting patterns highlight the tissue-specific functions of RBP1 in cancer biology. For endometrial cancer research, methodologies should include immunohistochemical analysis of RBP1 in endometrial cancer progression series (from normal to hyperplasia to carcinoma), correlation with hormone receptor status, and functional studies in endometrial cell lines with manipulated RBP1 expression.

Can RBP1 serve as a biomarker for cancer diagnosis or prognosis?

The biomarker potential of RBP1 varies by cancer type and requires careful evaluation: (1) in OSCC, higher RBP1 expression correlates with malignant features, suggesting its value as a prognostic marker ; (2) in endometrial cancer, loss of RBP1 expression may serve as a diagnostic indicator of malignant transformation ; (3) ELISA kits with sensitivity of 0.188 ng/ml and detection range of 0.313-20 ng/ml are available for quantifying RBP1 in human samples, facilitating potential clinical application ; (4) the tissue-specific expression patterns necessitate cancer-type-specific biomarker development; and (5) RBP1 might be most valuable as part of a multi-marker panel rather than as a standalone biomarker. Researchers evaluating RBP1's biomarker potential should conduct ROC curve analysis to determine sensitivity and specificity, Kaplan-Meier survival analysis to assess prognostic value, and multivariate analysis to determine independence from established prognostic factors.

What are the optimal methods for detecting RBP1 protein in experimental samples?

Several complementary methods can effectively detect RBP1 protein in experimental samples: (1) Western blotting using validated antibodies such as ab154881 (effective at 1/5000 dilution) for quantitative assessment of RBP1 protein levels, with the expected band size of 16 kDa ; (2) immunohistochemistry for tissue localization, with antibodies like ab154881 demonstrated to work effectively at 1/100 dilution for paraffin-embedded tissues ; (3) immunofluorescence for subcellular localization and co-localization studies, with ab154881 effective at 1/500 dilution after methanol fixation ; (4) ELISA for precise quantification in biological fluids, with commercial kits available that can detect RBP1 with sensitivity of 0.188 ng/ml ; and (5) mass spectrometry for unbiased detection and identification of post-translational modifications. When implementing these methods, researchers should include appropriate positive controls (liver tissue or cell lines known to express RBP1) and negative controls (antibody omission, isotype controls, or RBP1-depleted samples).

What genetic approaches are most effective for manipulating RBP1 expression?

Several genetic approaches have proven effective for manipulating RBP1 expression in experimental models: (1) transient overexpression via plasmid transfection, which has been successfully used in OSCC cell lines ; (2) siRNA-mediated knockdown, which effectively reduces RBP1 expression and has demonstrated functional consequences in cancer models ; (3) stable overexpression or knockdown using lentiviral or retroviral vectors for long-term studies; (4) CRISPR-Cas9 gene editing for complete knockout or targeted modifications of the RBP1 gene; and (5) inducible expression systems that allow temporal control over RBP1 expression. When manipulating RBP1 expression, researchers should verify changes at both mRNA (qRT-PCR) and protein (western blot) levels, include appropriate controls (empty vector, scrambled siRNA), and assess functional outcomes using assays appropriate to the research question. For cancer studies, these typically include proliferation, colony formation, migration, invasion, and in vivo tumor formation assays.

What cell and animal models are most appropriate for RBP1 research?

Several experimental models have been validated for RBP1 research: (1) OSCC cell lines including SCC15, SCC25, SCC9, and CAL27, with SCC15 showing particularly high RBP1 expression and Human Oral Keratinocyte (HOK) cells serving as normal controls ; (2) xenograft mouse models using RBP1-manipulated cell lines, which have demonstrated the impact of RBP1 silencing on tumor formation in vivo ; (3) primary cell cultures from patient samples, which maintain more physiological levels of RBP1 and related proteins; (4) 3D organoid cultures that better recapitulate tissue architecture for studying RBP1's role in differentiation; and (5) genetically modified mouse models for studying systemic effects of RBP1 alterations. When selecting models, researchers should consider the specific research question; for basic molecular interactions, simpler cell models may suffice, while complex physiological effects require animal models. For cancer studies, both gain-of-function and loss-of-function approaches should be employed to fully characterize RBP1's role.

How can researchers measure RBP1-mediated retinol transport activity?

Measuring RBP1-mediated retinol transport requires specialized approaches: (1) fluorescently labeled retinol analogs can track intracellular movement in live cells with normal versus altered RBP1 expression; (2) radiolabeled retinol uptake assays quantify transport efficiency and can be performed in cells with manipulated RBP1 levels; (3) retinol binding assays using purified RBP1 protein assess direct interactions and binding affinities; (4) FRET-based sensors can detect RBP1-retinol interactions in real-time within living cells; and (5) metabolic tracing with stable isotope-labeled retinol can follow conversion to downstream metabolites. Control experiments should include competitive inhibition with excess unlabeled retinol, comparison with cells lacking RBP1 expression, and parallel assessment of other retinol binding proteins to determine specificity. Subcellular fractionation followed by quantification of retinol in different compartments can provide spatial information about transport activity.

What proteomics approaches best identify RBP1 interaction partners?

Several proteomics approaches can effectively identify RBP1 interaction partners: (1) co-immunoprecipitation followed by mass spectrometry has successfully identified interactions such as the RBP1-CKAP4 axis ; (2) proximity-based labeling methods (BioID, APEX) can identify proteins in close proximity to RBP1 in living cells; (3) cross-linking mass spectrometry can capture transient interactions; (4) yeast two-hybrid screening can identify direct binary interactions; and (5) protein microarrays can screen for interactions with specific protein families. When analyzing potential interactions, researchers should implement stringent controls including reversed immunoprecipitation, IgG controls, and validation of key interactions using orthogonal methods such as co-immunoprecipitation followed by western blotting. Functional validation of identified interactions should include co-localization studies using immunofluorescence microscopy and genetic approaches to manipulate the expression of interaction partners.

How does post-translational modification affect RBP1 function in health and disease?

Post-translational modifications (PTMs) likely play crucial roles in regulating RBP1 function: (1) potential phosphorylation at serine/threonine residues may regulate RBP1's binding affinity for retinol or interaction with partner proteins like CKAP4; (2) ubiquitination could control RBP1 protein stability and turnover, influencing its availability in cells; (3) acetylation might affect RBP1's subcellular localization or binding properties; (4) cancer-specific modifications could contribute to RBP1's context-dependent roles in different tumor types; and (5) PTMs may serve as switches between RBP1's physiological function in retinol transport and its pathological role in processes like autophagy. Research methodologies should include mass spectrometry-based proteomics for comprehensive PTM mapping, phospho-specific antibodies, and site-directed mutagenesis of modification sites to determine their functional significance. The impact of PTMs on RBP1's interaction with CKAP4 represents a particularly promising research direction given the importance of this interaction in cancer.

What is the relationship between RBP1 expression and response to retinoid-based therapies?

The relationship between RBP1 expression and response to retinoid-based therapies represents an important clinical research question: (1) RBP1 levels may influence intracellular retinoid availability, potentially affecting response to exogenous retinoids; (2) in cancers with RBP1 overexpression, saturated binding capacity might alter therapeutic efficacy; (3) in cancers with RBP1 loss, reduced retinoid transport might compromise treatment response; (4) RBP1 expression could potentially serve as a predictive biomarker for retinoid therapy response; and (5) combination approaches targeting both RBP1 and retinoid signaling might overcome resistance mechanisms. Research approaches should include correlation of baseline RBP1 expression with clinical response to retinoid therapy, in vitro dose-response studies in cells with manipulated RBP1 levels, and analysis of RBP1 expression changes during treatment. Patient-derived xenograft models with varying RBP1 expression levels could evaluate treatment efficacy in a more physiologically relevant context.

How do epigenetic mechanisms regulate RBP1 expression in different tissues?

Epigenetic mechanisms likely play a significant role in regulating tissue-specific RBP1 expression: (1) DNA methylation of the RBP1 promoter region may contribute to silencing in some contexts and activation in others; (2) histone modifications could establish tissue-specific expression patterns during development; (3) chromatin remodeling complexes may alter accessibility of the RBP1 gene locus; (4) non-coding RNAs might post-transcriptionally regulate RBP1 mRNA stability or translation; and (5) these epigenetic mechanisms may explain the contrasting expression patterns of RBP1 in different cancer types. Research methodologies should include bisulfite sequencing to analyze DNA methylation patterns, ChIP-seq for histone modifications at the RBP1 locus, ATAC-seq to assess chromatin accessibility, and RNA-seq for non-coding RNA identification. Epigenetic editing using CRISPR-dCas9 fused to modifiers like DNMT3A or TET1 can establish causal relationships between specific epigenetic marks and RBP1 expression.

What are the emerging therapeutic strategies targeting the RBP1-CKAP4 axis?

Several emerging therapeutic strategies target the RBP1-CKAP4 axis: (1) small molecule inhibitors of the RBP1-CKAP4 protein-protein interaction could prevent downstream autophagy activation; (2) peptide-based inhibitors mimicking critical interaction domains could offer high specificity; (3) degraders (PROTACs) targeting RBP1 for proteasomal degradation in cancer contexts; (4) combination approaches with autophagy inhibitors could synergistically target this pathway; and (5) nanotechnology-based delivery systems could enhance targeting to tumor cells while sparing normal tissues. Research approaches should include high-throughput screening for interaction inhibitors, structure-based drug design targeting the RBP1-CKAP4 interface, preclinical evaluation in cell and animal models, and combination studies with established therapies. Importantly, therapeutic development must consider the essential physiological functions of RBP1 in retinoid transport to minimize adverse effects on normal tissues with high vitamin A requirements.

How does single-cell heterogeneity in RBP1 expression impact tumor biology?

Single-cell heterogeneity in RBP1 expression likely has significant implications for tumor biology: (1) subpopulations with varying RBP1 levels may exhibit different functional properties within the same tumor; (2) RBP1-high cells might possess enhanced migratory and invasive capabilities, contributing to metastatic potential; (3) differential autophagy activation across cell populations could create survival advantages under therapy; (4) RBP1 heterogeneity might contribute to tumor plasticity and adaptability to environmental challenges; and (5) spatial distribution of RBP1-expressing cells within the tumor microenvironment could influence interactions with stromal and immune cells. Research methodologies should include single-cell RNA-seq to profile RBP1 expression across tumor cell populations, spatial transcriptomics or multiplex immunofluorescence to map expression in the tissue context, and lineage tracing to track the fate of cells with different RBP1 expression levels. Functional studies isolating RBP1-high versus RBP1-low populations can determine their distinct contributions to tumor progression.

What are the key unresolved questions in RBP1 research?

Despite significant advances, several crucial questions about RBP1 remain unanswered: (1) the molecular mechanisms underlying its opposing roles in different cancer types (oncogenic in OSCC, tumor-suppressive in endometrial cancer) ; (2) the precise signaling pathway connecting RBP1-CKAP4 interaction to autophagy activation ; (3) whether targeting RBP1 or its interaction with CKAP4 represents a viable therapeutic strategy with acceptable safety profile; (4) the comprehensive protein interaction network of RBP1 beyond currently identified partners; and (5) how alterations in RBP1 expression affect global retinoid signaling across tissues. Addressing these questions will require integrative approaches combining structural biology, proteomics, functional genomics, and detailed mechanistic studies. The development of more sophisticated animal models and patient-derived systems will be essential for translational aspects of RBP1 research.

What methodological advances would accelerate progress in understanding RBP1 biology?

Several methodological advances could significantly accelerate progress in RBP1 research: (1) development of highly specific small molecule modulators of RBP1 function or protein interactions; (2) improved in vivo imaging techniques to visualize RBP1-mediated retinol transport in real-time; (3) more sophisticated tissue-specific and inducible genetic models to dissect context-dependent functions; (4) cryo-electron microscopy to resolve the structure of RBP1 protein complexes, particularly with CKAP4; and (5) advanced computational approaches to integrate multi-omics data for pathway analysis. These methodological advances would enable researchers to move beyond correlative observations to establish causal relationships between RBP1 alterations and disease phenotypes. Standardization of research protocols across laboratories would also facilitate more reliable cross-study comparisons and accelerate consensus building in the field.

Product Science Overview

Structure and Function

RBP1 is a small, soluble protein that binds retinol with high affinity. It is primarily found in the cytoplasm of cells and is involved in the intracellular transport of retinol. The protein ensures that retinol is delivered to specific target cells and tissues, where it can be metabolized into active forms such as retinoic acid. Retinoic acid is a potent regulator of gene expression and is involved in numerous developmental and physiological processes .

Expression and Production

Human recombinant RBP1 is typically produced using recombinant DNA technology. This involves inserting the gene encoding RBP1 into a suitable expression system, such as Escherichia coli (E. coli) or yeast, which then produces the protein. The recombinant protein is purified to high levels of purity, often exceeding 95%, to ensure its suitability for research and therapeutic applications .

Biological Importance

Retinol and its derivatives are vital for maintaining normal vision, immune function, and skin health. RBP1 plays a critical role in delivering retinol to the retina, where it is converted into retinal, a component of the visual pigment rhodopsin. Additionally, RBP1 is involved in the regulation of retinoic acid levels, which are crucial for embryonic development and cellular differentiation .

Clinical Relevance

Alterations in RBP1 expression or function can lead to various health issues. For instance, deficiencies in retinol transport can result in vitamin A deficiency, leading to conditions such as night blindness and an increased risk of infections. Conversely, excessive retinol levels can be toxic and cause hypervitaminosis A. Therefore, understanding the role of RBP1 in retinol metabolism is essential for developing therapeutic strategies for related disorders .

Research Applications

Human recombinant RBP1 is widely used in research to study retinol metabolism, retinoic acid signaling pathways, and their implications in health and disease. It serves as a valuable tool for investigating the molecular mechanisms underlying retinoid-related processes and for developing potential therapeutic interventions .

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.