CREG1 Human

Cellular Repressor of E1A-Stimulated Genes 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Biochemical Characteristics

CREG1 is a 220-amino acid glycoprotein with a molecular mass of 23–35 kDa post-translation . Key features include:

  • Domain Architecture: A split β-barrel fold with three N-linked glycosylation sites (N160, N193, N216 in humans) .

  • Dimerization: Forms homodimers via a hydrophobic interface, enhancing binding affinity to receptors like M6P/IGF2R .

  • Post-Translational Modifications:

    • Glycosylation in the endoplasmic reticulum, critical for secretion and lysosomal targeting .

    • Proteolytic processing by lysosomal cysteine proteases into smaller isoforms .

Lysosomal Biogenesis and Autophagy

  • Promotes lysosomal acidification and autophagosome-lysosome fusion via LAMP2 upregulation .

  • CRISPR/Cas9 knockout in mice exacerbates autophagy dysfunction, leading to cardiac hypertrophy in diabetic models .

Growth Suppression and Differentiation

  • Binds M6P/IGF2R to inhibit IGF-II-mediated proliferation .

  • Overexpression induces senescence in smooth muscle cells and embryonic carcinoma cells .

Key Protein Interactions

Interacting PartnerFunctional RoleMechanism
M6P/IGF2R Growth suppressionBlocks IGF-II endocytosis
Exocyst Sec8 Vesicle traffickingRegulates secretory pathways
FBXO27 Autophagy modulationStabilizes LAMP2 via ubiquitination

Clinical and Therapeutic Implications

Diabetic Cardiomyopathy

  • Transgenic CREG1 overexpression in mice improves cardiac function by 40% and reduces fibrosis via enhanced autophagy .

Vascular Remodeling

  • Adenovirus-mediated CREG1 delivery inhibits neointimal hyperplasia post-angioplasty by 60% .

Cancer

  • Downregulated in malignancies; recombinant CREG1 suppresses tumor growth in preclinical models .

Research Tools and Recombinant Protein Applications

  • Therapeutic Development: Recombinant CREG1 (e.g., 2380-CR-025 from R&D Systems) is being tested for proliferative disorders .

  • Experimental Use: E. coli-derived CREG1 (lacking glycosylation) facilitates structural studies .

Product Specs

Introduction
Cellular Repressor of E1A-Stimulated Genes 1 (CREG1) is a versatile protein that plays a dual role in regulating gene expression. It can activate or inhibit gene expression to influence cell proliferation and differentiation. One of its key functions is to counteract the transcriptional activation and cellular transformation induced by E1A. CREG1 exhibits partial sequence similarity with E1A and can bind to both the general transcription factor TBP and the tumor suppressor protein pRb in laboratory settings. Through these interactions, CREG1 contributes significantly to the intricate control of cell growth and differentiation.
Description
This product consists of the recombinant human CREG1 protein, produced in E. coli bacteria. It is a single polypeptide chain with a length of 213 amino acids (specifically, amino acids 32 to 220 of the full protein sequence) and a molecular weight of 23.6 kDa. To facilitate purification and detection, the protein has been engineered with a 24-amino acid His-tag attached to its N-terminus. Purification is carried out using proprietary chromatographic techniques, ensuring high purity.
Physical Appearance
Clear and colorless solution, sterilized by filtration.
Formulation
The provided CREG1 solution has a concentration of 1mg/ml. It is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 0.15M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the solution can be stored at 4°C. For longer storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advisable for long-term storage to maintain protein stability. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of the CREG1 protein is greater than 95%, as assessed by SDS-PAGE analysis.
Synonyms
Cellular repressor of E1A-stimulated genes 1, protein CREG1, CREG.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMRGGRDH GDWDEASRLP PLPPREDAAR VARFVTHVSD WGALATISTL EAVRGRPFAD VLSLSDGPPG AGSGVPYFYL SPLQLSVSNL QENPYATLTM TLAQTNFCKK HGFDPQSPLC VHIMLSGTVT KVNETEMDIA KHSLFIRHPE MKTWPSSHNW FFAKLNITNI WVLDYFGGPK IVTPEEYYNV TVQ

Q&A

What is the molecular structure of human CREG1?

Human CREG1 is a pyridoxine 5′-phosphate oxidase 2 domain-containing protein that exists as a tightly bound dimer. Each monomer forms a split β-barrel structure with a smooth, concave surface for dimerization and ragged areas that likely mediate protein-protein interactions. Approximately 20% of the CREG1 molecule's surface area participates in dimerization . The protein contains three N-glycosylation sites (N160, N193, and N216) exposed on the dimer surface, which may provide interfaces for protein interactions . Despite structural similarities to pyridoxine 5′-phosphate oxidase, CREG1's flavin mononucleotide (FMN) binding cleft is blocked, suggesting it lacks oxidoreductase activities .

How is CREG1 processed post-translationally?

CREG1 undergoes several key post-translational modifications:

  • Signal peptide cleavage: The protein contains a signal peptide (amino acids 1-31 in humans) that directs it to the endoplasmic reticulum and is subsequently cleaved .

  • N-glycosylation: After translation, CREG1 is modified by N-linked glycosylation in the endoplasmic reticulum. Treatment with peptide-N-glycosidase F significantly reduces CREG1's apparent molecular mass, confirming this modification .

  • Phosphorylation: Mass spectrometric analysis has confirmed that both human and mouse CREG1 contain phosphorylated N-glycans, including mannose-6-phosphate residues .

These modifications are critical for CREG1's proper localization and function, as deletion of the N-terminal signal peptide blocks both glycosylation and secretion .

What are the primary biological functions of CREG1?

CREG1 exhibits multiple biological functions across different cellular contexts:

  • Transcriptional regulation: Originally identified as a transcription repressor that antagonizes E1A oncoprotein-induced transcription and cellular transformation .

  • Differentiation control: When overexpressed or added as conditioned medium, CREG1 augments the differentiation of embryonal carcinoma cells .

  • Myogenesis regulation: CREG1 plays a crucial role in skeletal muscle satellite cell differentiation and muscle regeneration, with knockdown mice showing approximately 30% fewer newly formed fibers after injury compared to controls .

  • Cardiac protection: Functions as a myocardial protective factor, with its deficiency exacerbating cardiac dysfunction, hypertrophy, and fibrosis in diabetic cardiomyopathy models .

  • Tumor suppression: Treatment of PyMT tumor cells with recombinant CREG1 reduces proliferation, migration, and invasion, suggesting tumor-suppressive properties .

What are the optimal methods for studying CREG1 expression and localization?

Researchers investigating CREG1 expression and localization should consider a multi-modal approach:

  • Transcriptional analysis:

    • qRT-PCR for mRNA quantification

    • RNA-seq for comprehensive transcriptional profiling (as used in study GSE8479)

    • In situ hybridization for tissue-specific localization

  • Protein detection:

    • Western blotting with glycosylation-aware protocols (including PNGase F treatment as a control)

    • Immunoprecipitation for detection in conditioned media and protein interaction studies

    • Immunofluorescence microscopy with co-staining for endosomal/lysosomal markers to confirm subcellular localization

  • Glycosylation analysis:

    • Mass spectrometry to identify specific glycosylation patterns

    • Enzymatic deglycosylation assays to determine functional relevance of glycosylation

When studying CREG1 localization, it's essential to account for its distribution across multiple compartments (intracellular, membrane-associated, and secreted forms) and to use markers for endocytic-lysosomal compartments to accurately track its subcellular localization .

What genetic models are available for CREG1 functional studies?

Several genetic models have been developed for studying CREG1 function:

  • Knockout models:

    • Skeletal muscle mature myofibre-specific CREG1 knockout mice (myoblast/CREG1MKO)

    • Cardiac-specific CREG1 knockout mice

    • Global CREG1 knockout models (with developmental considerations)

  • Overexpression models:

    • CREG1 transgenic mice

    • Skeletal muscle satellite cells-specific CREG1 overexpression mice

    • Adeno-associated virus (AAV)-mediated CREG1 overexpression

  • In vitro models:

    • CREG1 knockdown in cell lines using siRNA or shRNA approaches

    • Plasmid-based overexpression systems using lipofectamine transfection

    • CRISPR/Cas9 gene editing for precise functional studies

When selecting a model, researchers should consider tissue specificity requirements and whether transient or stable modification is needed. For muscle regeneration studies, the cardiotoxin-induced injury model in tibialis anterior muscle has proven effective for assessing CREG1's role in myogenesis .

How does CREG1 regulate autophagy in diabetic cardiomyopathy?

CREG1 plays a critical role in regulating autophagy in the context of diabetic cardiomyopathy through several mechanisms:

  • LAMP2 regulation: CREG1 inhibits LAMP2 protein degradation by suppressing F-box protein 27 (FBXO27) expression. When CREG1 is knocked down, increased FBXO27 leads to enhanced LAMP2 degradation, thereby impairing autophagy .

  • Autophagy flux maintenance: In diabetic cardiomyopathy models, CREG1 deficiency exacerbates autophagy dysfunction, while CREG1 overexpression improves autophagy and subsequently cardiac function .

  • Cardiomyocyte protection: In palmitate-induced cardiomyocyte models mimicking diabetic conditions, CREG1 overexpression inhibits hypertrophy and improves autophagy, whereas CREG1 knockdown has opposite effects .

Methodologically, researchers investigating this pathway should:

  • Employ fluorescent LC3 reporters to monitor autophagy flux

  • Use co-immunoprecipitation to study the CREG1-FBXO27-LAMP2 interaction

  • Implement rescue experiments with LAMP2 overexpression to confirm the pathway's causality

  • Consider metabolic stress conditions (palmitate treatment, high glucose) relevant to diabetic environments

What is the relationship between CREG1 and AMPKα1 signaling in muscle regeneration?

CREG1 positively regulates muscle regeneration through the AMPKα1 signaling pathway:

  • CREG1 deficiency inhibits AMPKα1 signaling through C-CBL E3-ubiquitin ligase-mediated AMPKα1 degradation. This degradation occurs through K48-linked polyubiquitination of AMPKα1 at the K396 residue .

  • The regulatory relationship appears bidirectional, as CREG1 knockdown mice show approximately 30% reduction in newly formed muscle fibers after cardiotoxin injection, while muscle satellite cell-specific CREG1 overexpression mice exhibit approximately 20% more newly formed fibers .

  • RNA-seq analysis revealed that CREG1 deletion in impaired muscles leads to upregulation of inflammation and DKK3 expression, suggesting multiple downstream pathways .

For researchers studying this pathway, recommended approaches include:

  • Ubiquitination assays to detect K48-linked polyubiquitination of AMPKα1

  • Site-directed mutagenesis of AMPKα1 K396 to confirm the specific ubiquitination site

  • AAV-mediated C-CBL silencing in CREG1-deficient models to validate the pathway

  • Phosphorylation analysis of AMPKα1 and downstream targets

How does cathepsin B regulate CREG1 in the tumor microenvironment?

Cathepsin B (CTSB) negatively regulates CREG1 levels in the tumor microenvironment through proteolytic degradation:

  • Secretome analysis from tumor-macrophage co-cultures reveals an inverse correlation between CTSB expression and CREG1 abundance - high CREG1 in CTSB knockout conditions and low CREG1 in CTSB overexpressing conditions .

  • Direct evidence shows that cathepsin B can cleave CREG1 in vitro, suggesting proteolytic regulation .

  • This regulatory relationship appears functionally significant, as CREG1 acts as a tumor suppressor by reducing proliferation, migration, and invasion of tumor cells .

  • The cathepsin B-CREG1 axis represents a key component of tumor-stroma interaction in breast cancer models .

Researchers investigating this relationship should consider:

  • Proteomics analysis of tumor interstitial fluid for identifying cathepsin targets

  • In vitro cleavage assays with purified cathepsin B and CREG1

  • Mass spectrometry to identify specific CREG1 cleavage sites

  • Validation through genetic manipulation of CTSB in tumor-stroma co-culture systems

  • Orthotopic transplantation models to confirm in vivo relevance

What is the potential role of CREG1 in sarcopenia and age-related muscle dysfunction?

Transcriptional profiling of skeletal muscle biopsies from healthy older (N=25) and younger individuals revealed CREG1 association with human sarcopenia . This connection between CREG1 and age-related muscle deterioration suggests several research directions:

  • Age-related CREG1 expression patterns: Analyzing changes in CREG1 expression, localization, and post-translational modifications across the lifespan in muscle tissue.

  • Satellite cell function: Investigating whether age-related declines in muscle regenerative capacity involve CREG1 dysregulation in satellite cells.

  • Therapeutic potential: Exploring whether CREG1 supplementation or enhancement strategies might improve muscle regeneration in aged individuals.

  • Exercise responsiveness: Determining if exercise-induced muscle adaptations involve CREG1 regulation and whether this pathway is impaired in aging.

Methodologically, researchers should consider combining human biopsy analyses with age-appropriate animal models and interventional studies to establish causality between CREG1 alterations and sarcopenic phenotypes.

How might CREG1-targeted approaches be developed for therapeutic purposes?

Based on CREG1's multifunctional roles, several therapeutic strategies could be developed:

  • For muscle disorders:

    • AAV-mediated CREG1 gene delivery to enhance muscle regeneration

    • Small molecule inhibitors of C-CBL to prevent CREG1-mediated AMPKα1 degradation

    • Recombinant CREG1 protein therapy for acute muscle injuries

  • For diabetic cardiomyopathy:

    • CREG1 mimetics to improve cardiac autophagy

    • FBXO27 inhibitors to prevent LAMP2 degradation

    • Combination approaches targeting the CREG1-FBXO27-LAMP2 axis

  • For cancer treatment:

    • Recombinant CREG1 administration to suppress tumor growth

    • Cathepsin B inhibitors to prevent CREG1 degradation in the tumor microenvironment

    • CREG1 stabilizing compounds to enhance its tumor suppressive functions

When developing such approaches, researchers must consider:

  • CREG1's multiple post-translational modifications and their impact on function

  • Tissue-specific delivery methods to target relevant pathologies

  • Potential off-target effects given CREG1's involvement in multiple cellular processes

  • Optimal timing of intervention relative to disease progression

What are the key considerations when analyzing CREG1 glycosylation patterns?

CREG1 glycosylation analysis presents several technical challenges:

  • Heterogeneity: Human CREG1 contains three N-glycosylation sites (N160, N193, and N216) that may be differentially modified depending on cell type, developmental stage, or disease state .

  • Functional significance: These glycosylation sites are exposed on the dimer surface and may mediate interactions with receptors like M6P/IGF2R, influencing CREG1's localization and function .

  • Analytical approaches should include:

    • Site-directed mutagenesis of individual glycosylation sites

    • Enzymatic deglycosylation with PNGase F followed by functional assays

    • Lectin blotting to characterize glycan composition

    • Mass spectrometry for detailed glycan structure analysis

  • Experimental design considerations:

    • Cell-type specificity of glycosylation machinery

    • Secreted versus intracellular CREG1 may have different glycosylation patterns

    • Disease states may alter glycosylation machinery and patterns

Researchers should systematically address these aspects to fully understand how glycosylation influences CREG1's multifaceted biological functions.

How can researchers effectively study CREG1 in the context of protein-protein interactions?

Given CREG1's multiple interaction partners and cellular functions, studying its protein-protein interactions requires specialized approaches:

  • Identification methods:

    • Yeast two-hybrid screening (as used in the original CREG1 discovery)

    • Immunoprecipitation followed by mass spectrometry

    • Proximity labeling approaches (BioID, APEX)

    • Split-protein complementation assays

  • Validation approaches:

    • Co-immunoprecipitation under various cellular conditions

    • FRET/BRET to confirm direct interactions in living cells

    • Surface plasmon resonance for binding kinetics

    • Structural studies of complexes

  • Functional analysis:

    • Competitive binding assays to map interaction domains

    • Mutagenesis of key residues identified in structural studies

    • Cellular assays with interaction-deficient mutants

  • Context considerations:

    • Dimeric state of CREG1 may influence interaction profiles

    • Glycosylation status may affect protein-protein interactions

    • Subcellular localization (lysosomal versus secreted) determines accessible partners

By combining these approaches, researchers can build comprehensive interaction networks for CREG1 and identify key regulatory nodes for potential therapeutic targeting.

What emerging technologies could advance our understanding of CREG1 biology?

Several cutting-edge technologies offer promising avenues for deepening our understanding of CREG1:

  • Single-cell approaches:

    • Single-cell RNA-seq to identify cell populations with differential CREG1 expression

    • Single-cell proteomics to track CREG1 protein levels and modifications

    • Spatial transcriptomics to map CREG1 expression in complex tissues

  • Advanced imaging:

    • Super-resolution microscopy for detailed subcellular localization

    • Live-cell imaging with fluorescently tagged CREG1 to track trafficking

    • Correlative light and electron microscopy for ultrastructural context

  • Genome editing:

    • CRISPR/Cas9 screens to identify CREG1 regulators and effectors

    • Base editing for precise modification of glycosylation sites

    • Knock-in reporter systems for endogenous CREG1 tracking

  • Structural biology:

    • Cryo-EM studies of CREG1 in complex with interacting proteins

    • Hydrogen-deuterium exchange mass spectrometry for dynamic structural analysis

    • AlphaFold2 and other AI approaches for interaction prediction

These technologies, particularly when used in combination, could resolve outstanding questions about CREG1's regulation, trafficking, and context-specific functions.

How might CREG1 function across different tissues be integrated into a systems biology framework?

CREG1 operates in multiple tissues and cellular contexts, necessitating a systems biology approach:

  • Multi-omics integration:

    • Comparative transcriptomics across tissues expressing CREG1

    • Proteomics to identify tissue-specific interaction partners

    • Metabolomics to correlate CREG1 levels with metabolic states

    • Epigenomic analysis of CREG1 regulation across tissues

  • Network modeling:

    • Construction of CREG1-centered interactomes for different tissues

    • Pathway enrichment analysis for tissue-specific functions

    • Network perturbation simulations to predict intervention outcomes

    • Integration with existing pathway databases

  • Temporal considerations:

    • Developmental trajectories of CREG1 expression and function

    • Aging-related changes in CREG1 networks

    • Acute versus chronic response patterns

  • Physiological integration:

    • Cross-talk between CREG1 functions in muscle, heart, and other tissues

    • Hormonal and metabolic regulation of CREG1 across tissues

    • Whole-organism phenotyping of CREG1 models

This integrated approach could help resolve seemingly contradictory findings and identify key nodes for therapeutic targeting across multiple disease contexts.

Product Science Overview

Discovery and Function

CREG1 was first discovered due to its interaction with the adenovirus E1A oncoprotein. The E1A protein is known for its role in transforming cells and activating transcription. CREG1 counteracts these effects, thereby acting as a cellular repressor. This discovery highlighted the potential of CREG1 as a therapeutic target for diseases involving abnormal cell growth and transformation .

Role in Vascular Remodeling

One of the critical functions of CREG1 is its involvement in vascular remodeling. Studies have shown that CREG1 is a lysosomal glycoprotein that helps maintain vascular homeostasis. It has been observed that CREG1 gene expression is significantly decreased in remodeled vascular tissues, particularly in response to angiotensin II (Ang II). This downregulation is specific to Ang II and independent of blood pressure . The reduction in CREG1 expression leads to vascular hypertrophy and fibrosis, which are characteristic features of hypertensive vascular remodeling .

Tumor Suppression

CREG1 also exhibits tumor-suppressive properties. It has been shown to inhibit invasive cell growth and reduce the proliferation, migration, and invasion of tumor cells. This effect is particularly evident in the context of breast cancer, where CREG1 levels are negatively regulated by cathepsin B, a lysosomal protease. The overexpression of cathepsin B leads to a decrease in CREG1 levels, promoting tumor growth and invasion. Conversely, the inhibition or deletion of cathepsin B results in increased CREG1 levels, thereby suppressing tumor progression .

Therapeutic Potential

Given its role in vascular remodeling and tumor suppression, CREG1 holds significant therapeutic potential. It may serve as a target for interventions aimed at preventing hypertensive vascular diseases and certain types of cancer. The recombinant form of CREG1 has been studied for its protective effects against angiotensin II-induced hypertension and myocardial fibrosis, further underscoring its therapeutic relevance .

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.