UBE2G Human

Ubiquitin-Conjugating Enzyme E2G Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to UBE2G2 (Ubiquitin-Conjugating Enzyme E2 G2)

UBE2G2, encoded by the UBE2G2 gene on human chromosome 21q22.3, is a class I ubiquitin-conjugating enzyme (E2) critical for protein degradation pathways. It facilitates the transfer of ubiquitin to substrate proteins, targeting them for proteasomal degradation via endoplasmic reticulum-associated degradation (ERAD) and other processes. The enzyme shares 100% sequence identity with its murine ortholog and is ubiquitously expressed, with high levels in adult muscle .

Core Architecture

UBE2G2 adopts a conserved ubiquitin-fold structure:

  • Domain: Single domain with an antiparallel β-sheet (β1–β4), five α-helices (α1–α5), and two 3₁₀-helices (η1–η2) .

  • Active Site: Cysteine 89 (Cys89) is the catalytic residue, located near η1 .

  • Molecular Weight: 18.6 kDa (165 amino acids) .

E3 Ligase Partners

UBE2G2 collaborates with RING-type E3 ligases, including:

  • HRD1 (Hrd1): Mediates ERAD by targeting misfolded proteins .

  • TEB4: Involved in ubiquitination of specific substrates .

  • CRL4 CRBN: Indirectly implicated in cereblon-mediated degradation via redundancy with UBE2G1 .

Mechanistic Role

ProcessUBE2G2 FunctionKey Substrates/Pathways
ERADK48-linked polyubiquitinationMisfolded ER proteins
CRL4 CRBN NeomorphicRedundant role with UBE2G1IKZF1, GSPT1 (in IMiD therapy)
Substrate UbiquitinationRequires E3 proximity for processivityCdt1 (via CRL4 Cdt2)

Cooperative E2 Activity:

  • UBE2D3 and UBE2G1: Sequentially catalyze K48-linked polyubiquitination in cereblon pathways .

  • UBE2G2 Redundancy: Partially compensates for UBE2G1 loss in substrate degradation .

Key Studies

Study (Year)MethodologyFindingsImplications
Arai et al. (2006)X-ray crystallography (2.56 Å)Structural similarity to yeast Ubc7; C-terminal helix stabilizes E3 binding Basis for E2–E3 interaction specificity
Choi et al. (2015)In vitro ubiquitination assaysUBE2G1/2 mediate K48-linked polyubiquitination for proteasomal degradation Targeting UBE2G2 in therapeutic strategies
Schafer et al. (2018)CRISPR knockout in myeloma cellsUBE2G1 loss confers resistance to lenalidomide; UBE2G2 redundancy observed Patient stratification for cereblon modulators

Therapeutic Potential

  • BioPROTACs: Human E2 enzymes (e.g., UBE2B, UBE2D1) are repurposed for targeted protein degradation .

  • Cereblon Modulators: UBE2G2 may influence resistance to lenalidomide/pomalidomide in myeloma .

Product Specs

Introduction
Protein modification with ubiquitin is a crucial cellular process for marking abnormal or short-lived proteins for degradation. This process involves at least three types of enzymes: ubiquitin-activating enzymes (E1s), ubiquitin-conjugating enzymes (E2s), and ubiquitin-protein ligases (E3s). Ubiquitin-Conjugating Enzyme E2G (UBE2G1), a member of the E2 ubiquitin-conjugating enzyme family, facilitates the attachment of ubiquitin to other proteins. UBE2G1 is specifically involved in the breakdown of muscle-specific proteins.
Description
Recombinant human UBE2G, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 193 amino acids (1-170 a.a.) with a molecular weight of 21.9 kDa. This protein is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The UBE2G protein solution (0.5mg/ml) is supplied in phosphate buffered saline (pH 7.4) containing 30% glycerol and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
E217K, UBC7, UBE2G, Ubiquitin-conjugating enzyme E2 G1, E2 ubiquitin-conjugating enzyme G1, E217K, UBC7, Ubiquitin carrier protein G1, Ubiquitin-protein ligase G1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMTELQSA LLLRRQLAEL NKNPVEGFSA GLIDDNDLYR WEVLIIGPPD TLYEGGVFKA HLTFPKDYPL RPPKMKFITE IWHPNVDKNG DVCISILHEP GEDKYGYEKP EERWLPIHTV ETIMISVISM LADPNGDSPA NVDAAKEWRE DRNGEFKRKV ARCVRKSQET AFE.

Q&A

What is UBE2G and what is its role in the ubiquitination pathway?

UBE2G (Ubiquitin-Conjugating Enzyme E2 G) is a family of E2 enzymes in humans that function within the ubiquitin-proteasome system. The most well-characterized member, UBE2G2 (also known as UBC7 in other species), acts as an essential component of the endoplasmic reticulum-associated degradation (ERAD) pathway, which identifies and eliminates misfolded proteins in the endoplasmic reticulum. UBE2G2 works in conjunction with specific E3 ubiquitin ligases to assemble K48-linked polyubiquitin chains on substrate proteins, marking them for subsequent degradation by the 26S proteasome . This process is critical for cellular protein quality control and homeostasis. Unlike many other E2 enzymes, UBE2G2 has been shown to have the specialized ability to preassemble polyubiquitin chains on its active site cysteine before transferring them to target substrates in a process dependent on its interaction with cognate E3 ligases .

How was UBE2G initially identified and characterized in humans?

UBE2G was first isolated from a human fetal-brain cDNA library as a novel ubiquitin-conjugating enzyme. The initially identified cDNA, designated UBE2G, contained an open reading frame of 510 nucleotides encoding a protein of 170 amino acids. The predicted peptide product demonstrated 74% identity at the amino acid level with UBC7 of C. elegans and significant homology with UBC7s of other species . Northern blot analysis revealed strong expression patterns with 4.4-kb, 2.4-kb, and 1.6-kb transcripts predominantly in skeletal muscle, while weaker expression was observed in 15 other examined tissues. Through radiation-hybrid mapping techniques, researchers localized this gene to chromosome band 1q42 . This initial characterization established UBE2G as a distinct human member of the evolutionarily conserved E2 family with tissue-specific expression patterns.

What is the structural organization of UBE2G2 and how does it relate to function?

UBE2G2's structure consists of a conserved ubiquitin-conjugating (UBC) fold that characterizes all E2 enzymes. Nuclear magnetic resonance (NMR) spectroscopy has revealed that while the core structure aligns well with crystallographic structures of other E2 enzymes, UBE2G2 possesses unique catalytically important loops (residues 95-107 and 130-135) that flank the active site cysteine . These loops are poorly defined in structural studies and exhibit high dynamics in solution, as demonstrated by 15N spin relaxation and residual dipolar coupling analysis .

The structural data suggests that UBE2G2 requires interaction with protein partners (E3 ligases, acceptor ubiquitin substrate, or thiolester-linked donor ubiquitin) to adopt its catalytically relevant conformation. Within the NMR structural ensemble, interactions were observed between His94 and the highly mobile loop residues Asp98 and Asp99, indicating a potential mechanism where His94 functions as a general base activated by the carboxylate side-chains of these aspartic acid residues . This structural arrangement is critical for UBE2G2's specialized function in the ERAD pathway.

What are the most effective techniques for studying UBE2G2 structure and dynamics?

Several complementary techniques have proven valuable for investigating UBE2G2's structure and dynamics. Nuclear magnetic resonance (NMR) spectroscopy stands out as particularly effective for studying UBE2G2, as demonstrated in comprehensive structural studies . NMR allows researchers to examine both the solution structure and the backbone dynamics, revealing critical information about flexible regions that may be poorly resolved in crystal structures. For studying UBE2G2:

For functional studies, in vitro ubiquitination assays with purified components (E1, UBE2G2, E3 ligases, and substrates) remain the gold standard for biochemical characterization .

How can researchers effectively identify E3 ligase partners for UBE2G2?

Identifying E3 ligase partners for UBE2G2 requires a multi-faceted approach combining both screening techniques and validation methods. Based on comprehensive studies of E2-E3 interactions , researchers should consider:

  • Yeast two-hybrid (Y2H) screening: A global Y2H screen has proven effective for mapping interactions between E2s and RING-type E3s. This approach has uncovered numerous high-quality E2-E3 interactions, including many involving UBE2G2 . When designing Y2H experiments:

    • Use the UBC-fold of UBE2G2 as bait

    • Screen against a library of RING domains from potential E3 partners

    • Include appropriate controls (known interactors and non-interactors)

    • Validate positive hits with secondary reporter assays

  • In vitro protein interaction assays: Direct biochemical validation of Y2H hits using methods such as:

    • GST pulldown assays with recombinant proteins

    • Surface plasmon resonance (SPR) to determine binding kinetics

    • Isothermal titration calorimetry (ITC) for thermodynamic parameters of binding

  • Functional ubiquitination assays: Testing whether identified E3s can stimulate UBE2G2's ubiquitin transfer activity in reconstituted systems.

  • Cellular validation: Co-immunoprecipitation of endogenous proteins or proximity labeling approaches (BioID, APEX) to confirm interactions in cellular contexts.

A comprehensive E2-E3 interaction study revealed that several E3 ligases interact with UBE2G2, including gp78 (autocrine motility factor receptor), parkin, HRD1, and TEB4 . The correlation between physical interactions identified in Y2H screens and functional E2-E3 pairs in in vitro ubiquitination experiments was found to be high, suggesting that physical interaction screening is a reliable predictor of functional partnerships .

What experimental systems are optimal for studying UBE2G2's role in ERAD?

Several experimental systems have been established for investigating UBE2G2's role in endoplasmic reticulum-associated degradation (ERAD):

  • Cell-based ERAD reporter systems:

    • Cells expressing well-characterized ERAD substrates (e.g., TCR-α, CD3-δ, CYP3A4) tagged with fluorescent or luminescent markers to monitor degradation rates

    • CRISPR/Cas9-based genetic screening approaches using HLA-I downregulation as a readout, as demonstrated in studies of US2-mediated ERAD

    • Flow cytometry-based assays measuring surface expression of membrane proteins processed through ERAD

  • Reconstituted in vitro ERAD systems:

    • Purified components (E1, UBE2G2, E3 ligases like gp78 or HRD1, and model substrates)

    • ER-derived microsomes supplemented with cytosolic factors

    • These systems allow for mechanistic studies of ubiquitin chain assembly and transfer

  • Cell-free retrotranslocation assays:

    • Semi-permeabilized cells or isolated ER membranes containing ERAD substrates

    • Addition of cytosolic factors including UBE2G2 and interacting partners

    • Monitoring substrate extraction from the ER membrane

  • Genetic complementation systems:

    • UBE2G2 knockout cell lines rescued with wild-type or mutant UBE2G2 variants

    • This approach allows structure-function analysis of UBE2G2 domains and residues

For studying UBE2G2 in viral subversion of ERAD, systems using HCMV US2 expression have proven particularly informative. In one study, U937 monocytic cells stably expressing an HLA-A2 molecule with an N-terminal eGFP tag were generated, allowing monitoring of HLA-I expression levels by flow cytometry . Upon stable introduction of HCMV US2, the chimeric HLA-I molecules and endogenous HLA-I proteins were efficiently degraded. This system, combined with CRISPR/Cas9 targeting of UBE2G2, demonstrated UBE2G2's essential role in US2-mediated HLA-I downregulation .

How does UBE2G2 contribute to protein quality control in the endoplasmic reticulum?

UBE2G2 plays a central role in the endoplasmic reticulum-associated degradation (ERAD) pathway, which is critical for maintaining ER homeostasis by eliminating misfolded or unassembled proteins. As a specialized E2 ubiquitin-conjugating enzyme within this pathway, UBE2G2 contributes to protein quality control through several mechanisms:

  • K48-linked polyubiquitin chain assembly: UBE2G2 has the specialized ability to assemble K48-linked polyubiquitin chains, which serve as the primary signal for targeting substrates to the 26S proteasome. Unlike many E2 enzymes that transfer single ubiquitin moieties, UBE2G2 can preassemble these chains on its active site cysteine before transferring them to substrates in a process dependent on cognate E3 ligases .

  • Collaboration with multiple ERAD E3 ligases: UBE2G2 works with several membrane-bound E3 ubiquitin ligases including:

    • gp78 (also known as autocrine motility factor receptor)

    • HRD1

    • TEB4 (also known as MARCH6)

    • Parkin
      These E3 partners provide substrate specificity and facilitate the transfer of ubiquitin from UBE2G2 to various ERAD targets .

  • Processing diverse ERAD substrates: Through its E3 partners, UBE2G2 mediates the ubiquitination and subsequent degradation of various substrates including:

    • T-Cell Receptor α subunit (TCR-α)

    • CD3 δ subunit (CD3-δ)

    • Human liver cytochrome P450 CYP3A4

    • Pael receptor

    • Inositol 1,4,5-trisphosphate receptors

  • Coordinating retrotranslocation and degradation: As ubiquitin is not found in the ER lumen, target proteins identified by the ER quality control system must be retrotranslocated to the cytoplasm for ubiquitination and degradation. UBE2G2's activity is coordinated with the retrotranslocation machinery to ensure efficient coupling of these processes .

Dysfunctions in this UBE2G2-dependent quality control system have been implicated in various diseases, including those associated with protein misfolding such as certain neurodegenerative disorders and cystic fibrosis .

What is known about UBE2G2's role in viral immune evasion mechanisms?

UBE2G2 has been identified as a critical component in viral immune evasion mechanisms, particularly in the context of human cytomegalovirus (HCMV) infection. Research has revealed that UBE2G2 plays an essential role in the US2-mediated degradation of human leukocyte antigen class I (HLA-I) molecules, which represents a key viral strategy to evade immune surveillance:

  • US2-mediated HLA-I degradation pathway: During HCMV infection, the viral protein US2 targets HLA-I molecules for degradation, preventing the presentation of viral antigens to cytotoxic T cells. This process occurs through the ERAD pathway and requires the coordinated action of specific cellular components .

  • UBE2G2 as an essential factor: Through a CRISPR/Cas9-based library targeting all known human E2 enzymes, UBE2G2 was identified as an essential E2 enzyme for US2-mediated HLA-I downregulation. Upon UBE2G2 depletion, HLA-I molecules were rescued from degradation in US2-expressing cells .

  • Mechanistic insights: When UBE2G2 is depleted, HLA-I molecules accumulate in an ER-resident complex containing both US2 and the E3 ligase TRC8. This suggests that ubiquitination mediated by UBE2G2 is required for extraction of HLA-I molecules from the ER and their subsequent degradation .

  • Specificity of UBE2G2 function: While another E2 enzyme, UBE2D3, was also found to contribute to US2-mediated HLA-I degradation, UBE2G2 showed the strongest effect in rescue experiments. The specificity of this function was confirmed by the observation that targeting the UBE2G2 homolog UBE2G1 did not affect HLA-I expression .

  • Counteracting factors: Interestingly, the same screening approach identified UBE2J2 as a counteracting E2 enzyme, depletion of which further downregulated HLA-I in US2-expressing cells, suggesting a complex regulatory network .

This research highlights UBE2G2's role in viral pathogenesis and immune evasion, suggesting it could be a potential therapeutic target for modulating immune responses during viral infections.

How is UBE2G2 expression regulated in different human tissues?

UBE2G2 exhibits distinct expression patterns across human tissues, suggesting tissue-specific regulation and functional requirements:

  • Predominant expression in skeletal muscle: Northern blot analysis has revealed strong expression of UBE2G transcripts (4.4-kb, 2.4-kb, and 1.6-kb) in skeletal muscle, indicating a potentially significant role in muscle protein homeostasis .

  • Broader tissue distribution: While skeletal muscle shows the highest expression, weaker UBE2G expression has been observed in 15 other examined tissues, suggesting a widespread but variable requirement for this E2 enzyme across the body .

  • Developmental regulation: The initial isolation of UBE2G from a human fetal-brain cDNA library indicates expression during developmental stages, which may reflect roles in neuronal development and protein quality control during brain formation .

  • Transcriptional regulation: The presence of multiple transcript sizes (4.4-kb, 2.4-kb, and 1.6-kb) suggests complex transcriptional and/or post-transcriptional regulation, potentially involving alternative promoter usage, splicing, or polyadenylation sites .

  • Correlation with ERAD substrate load: While not explicitly stated in the provided literature, it is reasonable to hypothesize that UBE2G2 expression levels may correlate with tissues that experience high rates of protein synthesis and folding in the ER, and consequently require robust ERAD capacity.

The tissue-specific expression pattern of UBE2G2, particularly its enrichment in skeletal muscle, may be relevant to understanding muscle-specific protein quality control mechanisms and potentially the pathophysiology of muscle diseases involving protein homeostasis dysregulation.

How do the dynamic loops of UBE2G2 contribute to its catalytic specificity?

The catalytic mechanism of UBE2G2 involves unique structural features, particularly two dynamic loops (residues 95-107 and 130-135) that flank the active site cysteine. These loops exhibit distinctive properties that contribute to UBE2G2's functional specificity:

  • Conformational flexibility: NMR studies have revealed that these two loops are highly dynamic in solution, as directly demonstrated by 15N spin relaxation and residual dipolar coupling analysis . This mobility contrasts with the well-defined structure of the E2 core, suggesting functional significance.

  • Catalytic activation mechanism: Within the NMR structural ensemble, interactions were observed between His94 and the highly mobile loop residues Asp98 and Asp99. This arrangement supports a potential mechanism where His94 functions as a general base activated by the carboxylate side-chains of these aspartic acid residues . This catalytic triad-like arrangement may facilitate:

    • Deprotonation of the active site cysteine

    • Nucleophilic attack on the E1-ubiquitin thioester

    • Enhanced reactivity during polyubiquitin chain formation

  • Partner-induced conformational changes: The structural data suggests that UBE2G2 requires interaction with protein partners (E3 ligases, acceptor ubiquitin substrate, or thiolester-linked donor ubiquitin) to assume its catalytically relevant conformation . The dynamic loops likely undergo induced-fit conformational changes upon binding to:

    • Position the active site optimally for catalysis

    • Accommodate different ubiquitin orientations during chain formation

    • Provide specificity for K48-linked chain assembly

  • Specificity for ERAD: The distinctive structural properties of these loops likely contribute to UBE2G2's specialized role in the ERAD pathway, potentially:

    • Facilitating interactions with membrane-bound E3 ligases

    • Enabling the preassembly of K48-linked polyubiquitin chains

    • Ensuring efficient transfer of preassembled chains to substrates

  • Evolutionary conservation: Comparative analysis of UBE2G2 across species would likely reveal conservation of key residues within these loops, particularly those involved in the proposed catalytic mechanism, highlighting their functional importance.

For researchers studying UBE2G2 catalysis, these dynamic loops represent critical structural elements that warrant detailed investigation through mutagenesis, structural studies of protein complexes, and computational modeling approaches to fully elucidate their contribution to catalytic specificity.

What are the determinants of E2-E3 specificity in UBE2G2 interactions?

Understanding the molecular basis of UBE2G2's selective interactions with specific E3 ligases is crucial for elucidating its function in the ubiquitination cascade. Several determinants contribute to this specificity:

  • Structural determinants of E2-E3 recognition:

    • UBE2G2, like other E2s, interacts with RING-domain E3 ligases primarily through its α1-helix, loop between α1 and β1, and loops 4 and 7

    • Specific residues within these regions create a recognition surface that determines E3 selectivity

    • The dynamic loops flanking the active site cysteine may provide additional interaction surfaces specific to UBE2G2's E3 partners

  • Connectivity patterns in E2-E3 interaction networks:

    • Comprehensive mapping of E2-E3 interactions has revealed that certain E2s, including UBE2G2, exhibit different degrees of "promiscuity" in their E3 interactions

    • UBE2G2 demonstrates a selective interaction pattern with specific RING E3 ligases such as gp78, parkin, HRD1, and TEB4

    • These interaction patterns reflect the specialized role of UBE2G2 in ERAD and related pathways

  • Biochemical validation of specificity:

    • Physical interactions identified in Y2H screens correlate well with functional E2-E3 pairs in in vitro ubiquitination experiments

    • This correlation validates the biological relevance of the observed interaction specificity

    • Mutagenesis studies have demonstrated that altering key residues in E2 enzymes can redirect their E3 specificity, as shown for UBE2N (Ubc13)

  • Subcellular localization as a determinant of interaction:

    • UBE2G2's interactions with E3 ligases are partially determined by their shared localization at the ER membrane

    • The primary E3 partners of UBE2G2 (gp78, HRD1, TEB4) are all ER-resident E3 ligases involved in ERAD

    • This co-localization facilitates their interactions and functional coupling in the ERAD pathway

  • Functional consequences of specificity:

    • Different UBE2G2-E3 pairs likely target distinct sets of substrates

    • The specific pairing of UBE2G2 with TRC8 in US2-mediated HLA-I degradation illustrates how these specific interactions are exploited in biological processes such as viral immune evasion

Understanding these determinants can guide experimental approaches for manipulating UBE2G2-E3 interactions and potentially developing therapeutic strategies targeting specific aspects of UBE2G2 function in disease contexts.

What experimental contradictions exist in UBE2G2 research and how might they be resolved?

Several areas of UBE2G2 research present apparent contradictions or knowledge gaps that warrant further investigation:

These contradictions represent opportunities for researchers to design experiments that address fundamental questions about UBE2G2 biology, potentially leading to new insights into ubiquitination mechanisms and protein quality control pathways.

What are the optimal conditions for expression and purification of active UBE2G2?

Producing high-quality, active UBE2G2 protein is essential for biochemical and structural studies. Based on established protocols in the field, researchers should consider the following optimization strategies:

  • Expression systems:

    • E. coli: The most common system for UBE2G2 expression, typically using BL21(DE3) or similar strains

    • Vector selection: pET-28b with a His-tag and TEV cleavage site has been successfully used for UBE2G2 expression

    • Fusion tags: N-terminal His6-tag facilitates purification while maintaining activity; GST or MBP tags may improve solubility but should be removable

  • Expression conditions:

    • Temperature: Lower temperatures (16-18°C) after induction often improve solubility

    • Induction: IPTG concentration of 0.4-0.5 mM is typically sufficient

    • Duration: Extended expression (overnight) at lower temperatures typically yields better results than short, high-temperature expression

  • Purification strategy:

    • Initial capture: Ni-NTA affinity chromatography with imidazole gradient elution

    • Tag removal: TEV protease cleavage followed by reverse Ni-NTA to remove the cleaved tag

    • Further purification: Size exclusion chromatography on Superdex 75 or similar matrix

    • Buffer conditions: Typically 20-50 mM Tris-HCl or phosphate buffer, pH 7.5-8.0, with 100-150 mM NaCl

  • Activity considerations:

    • Reducing agents: Include DTT (1-5 mM) or TCEP (0.5-1 mM) to maintain the active site cysteine in a reduced state

    • Storage: Flash-freeze in small aliquots with 10% glycerol and store at -80°C

    • Active site titration: Determine the fraction of active enzyme using ubiquitin-vinyl sulfone or similar active site probes

  • Quality control:

    • Homogeneity: Assess by SDS-PAGE, dynamic light scattering, and size exclusion chromatography

    • Activity assays: Thioester formation with ubiquitin in the presence of E1

    • Structural integrity: Circular dichroism spectroscopy or thermal shift assays

These optimization strategies should be tailored to the specific requirements of the planned experiments, with particular attention to maintaining the catalytic activity of the enzyme by protecting the active site cysteine from oxidation.

How can researchers effectively design mutational studies of UBE2G2?

Designing effective mutational studies requires careful consideration of UBE2G2's structure, function, and interaction networks. The following systematic approach is recommended:

  • Target selection based on structural information:

    • Catalytic residues: The active site cysteine (Cys89) is essential for ubiquitin conjugation and represents the primary target for catalytic studies

    • His94-Asp98/Asp99 network: These residues form a potential catalytic network and are prime targets for understanding the activation mechanism

    • Dynamic loops: Residues 95-107 and 130-135 are highly dynamic regions flanking the active site and crucial for function

    • E3-binding interface: The α1-helix and loops connecting secondary structure elements form the E3-binding surface

  • Types of mutations to consider:

    • Alanine scanning: Systematic replacement of residues with alanine to identify essential amino acids

    • Conservative substitutions: Replacing residues with similar amino acids to probe specific chemical properties (e.g., D→E, K→R)

    • Non-conservative substitutions: Changing charge, polarity, or size to dramatically alter properties

    • Cysteine mutations: For subsequent chemical modification or cross-linking studies

    • Mimicking post-translational modifications: Phosphomimetic mutations (S/T→D/E) if phosphorylation is suspected

  • Experimental validation approaches:

    • In vitro activity assays:

      • Thioester formation with ubiquitin

      • Free polyubiquitin chain assembly

      • E3-dependent substrate ubiquitination

    • Binding studies:

      • Surface plasmon resonance or isothermal titration calorimetry for E3 binding

      • Pulldown assays with potential interacting partners

    • Structural studies:

      • NMR to assess local structural changes and dynamics

      • Hydrogen-deuterium exchange mass spectrometry

    • Cellular studies:

      • Complementation of UBE2G2 knockout/knockdown cells

      • HLA-I degradation assays in the US2 system

  • Design of altered-specificity mutants:
    The approach used to alter the E3 interactions of UBE2N towards UBE2D2 specificity provides a template for UBE2G2 studies:

    • Identify key residues at the E3 interface through sequence alignment and structural analysis

    • Generate point mutations that might redirect E3 specificity

    • Test mutants in yeast two-hybrid and biochemical assays against a panel of E3s

  • Controls and validation:

    • Expression/stability controls: Ensure mutations don't simply destabilize the protein

    • Activity controls: Include wild-type UBE2G2 and catalytically dead (C89A) controls

    • Specificity controls: Test effects on interactions with multiple E3 partners

By following this structured approach, researchers can generate meaningful insights into UBE2G2 function while avoiding common pitfalls in mutational analysis.

What approaches can be used to study UBE2G2 in physiologically relevant contexts?

To understand UBE2G2 function in physiologically relevant settings, researchers should consider multi-layered approaches that bridge biochemical mechanisms with cellular and organismal contexts:

  • Cellular models with endogenous protein manipulation:

    • CRISPR/Cas9 gene editing:

      • Generate UBE2G2 knockout cell lines for loss-of-function studies

      • Create knock-in cell lines with tagged or mutant versions for localization and functional studies

      • Engineer conditional alleles (e.g., degron-tagged) for temporal control of UBE2G2 levels

    • Endogenous tagging approaches:

      • Tag endogenous UBE2G2 with fluorescent proteins or affinity tags

      • Use split-GFP or proximity labeling approaches to visualize interactions with E3 partners

    • Cell type selection:

      • Use cell types with high ERAD requirements (e.g., professional secretory cells)

      • Consider skeletal muscle cells given UBE2G2's high expression in muscle tissue

  • Physiological stress conditions:

    • ER stress induction:

      • Pharmacological stressors (tunicamycin, thapsigargin)

      • Expression of misfolding-prone proteins

      • Glucose deprivation or hypoxia

    • Viral infection models:

      • HCMV infection to study US2-mediated HLA-I degradation

      • Other viral systems that manipulate ERAD

    • Tissue-specific stress:

      • Exercise-induced stress in muscle cells

      • Differentiation-associated ER remodeling

  • Patient-derived materials:

    • Cells from patients with ERAD-associated disorders:

      • Cystic fibrosis

      • α1-antitrypsin deficiency

      • Certain neurodegenerative diseases

    • Genetic variation analysis:

      • Examine effects of naturally occurring UBE2G2 variants on function

      • Correlate UBE2G2 expression levels with disease progression

  • Advanced cellular techniques:

    • Live-cell imaging of ERAD dynamics:

      • Fluorescently labeled ERAD substrates

      • Optogenetic control of UBE2G2 activity

    • Single-cell analysis:

      • Correlate UBE2G2 levels with ERAD efficiency at single-cell resolution

      • Examine cell-to-cell variability in response to ER stress

    • Organoid and 3D culture systems:

      • Study UBE2G2 in more complex cellular environments

      • Tissue-specific ERAD regulation

  • Translational approaches:

    • Pharmacological modulation:

      • Screen for compounds that modulate UBE2G2 activity

      • Test effects of proteasome inhibitors on UBE2G2-dependent pathways

    • Therapeutic strategies:

      • Evaluate UBE2G2 as a potential target in diseases with ERAD dysfunction

      • Explore manipulation of UBE2G2-dependent pathways in viral infections

These approaches collectively enable researchers to connect the molecular mechanisms of UBE2G2 function to its physiological roles and potential involvement in disease states.

How does UBE2G2 compare functionally with other human E2 enzymes?

UBE2G2 possesses distinctive characteristics that set it apart from other human E2 enzymes in the ubiquitination cascade, while also sharing core functional features:

  • Functional specialization within the E2 family:

    • Among the 35 human E2 enzymes identified , UBE2G2 is specialized for functions in the ERAD pathway

    • UBE2G2 demonstrates a moderate degree of connectivity with E3 ligases compared to highly promiscuous E2s like UBE2D2 (UbcH5B) or highly selective E2s

    • While many E2s transfer single ubiquitin moieties, UBE2G2 has the specialized ability to preassemble K48-linked polyubiquitin chains on its active site cysteine

  • Comparison with core E2 enzymes:

    • UBE2D family (UbcH5): These highly versatile E2s interact with numerous E3s and participate in multiple cellular processes, while UBE2G2 is more specialized for ERAD

    • UBE2L3 (UbcH7): Unlike UBE2G2, UBE2L3 works exclusively with HECT and RBR E3 ligases, not RING E3s

    • UBE2N (Ubc13): Specialized for K63-linked chain formation (typically associated with signaling rather than degradation), contrasting with UBE2G2's K48 specificity

  • Linkage specificity:

    • UBE2G2 predominantly forms K48-linked polyubiquitin chains, which serve as the canonical degradation signal

    • This contrasts with E2s specialized for other linkage types:

      • UBE2N/UBE2V1: K63 linkages (signaling)

      • UBE2S: K11 linkages (cell cycle regulation)

      • UBE2K: K48 linkages (but with different mechanisms)

  • Structural comparison with other E2s:

    • All E2s share the conserved UBC fold, with the catalytic cysteine positioned similarly

    • UBE2G2's distinguishing features include its highly dynamic loops (residues 95-107 and 130-135) flanking the active site

    • The potential catalytic triad-like arrangement involving His94, Asp98, and Asp99 represents a specialized activation mechanism

  • Relative contribution to specific pathways:

    • In US2-mediated HLA-I degradation, UBE2G2 has a stronger effect than UBE2D3, while UBE2J2 counteracts this process

    • This demonstrates how different E2s can have opposing or complementary roles within the same biological pathway

This comparative perspective highlights UBE2G2's specialized niche within the human ubiquitination system, combining a conserved catalytic core with unique structural and functional adaptations tailored to its role in ERAD and related processes.

How is UBE2G2 function conserved or diversified across species?

UBE2G2 represents a conserved component of the ubiquitination machinery across diverse eukaryotic species, with both preserved core functions and species-specific adaptations:

  • Evolutionary conservation:

    • UBE2G2 is the human ortholog of the well-characterized yeast Ubc7 enzyme

    • The human UBE2G protein shows 74% amino acid identity with UBC7 of C. elegans and high homology with UBC7s of other species

    • This strong conservation suggests fundamental importance in eukaryotic protein quality control

  • Functional conservation in ERAD:

    • The role of UBE2G2/Ubc7 in ERAD pathways is preserved from yeast to humans

    • In both yeast and mammals, this E2 works with membrane-bound E3 ligases to target misfolded ER proteins

    • The specificity for K48-linked polyubiquitin chain formation is maintained across species

  • Species-specific adaptations:

    • Mammals have two UBE2G paralogs (UBE2G1 and UBE2G2) compared to the single Ubc7 in yeast

    • This duplication likely allowed functional diversification, as evidenced by the non-redundant functions of UBE2G1 and UBE2G2

    • Higher organisms show more complex regulation of UBE2G2 expression, including tissue-specific patterns not observed in unicellular organisms

  • E3 interaction network evolution:

    • The E3 interaction network has expanded considerably from yeast to humans

    • While yeast Ubc7 primarily interacts with the E3 ligase Hrd1, human UBE2G2 works with multiple E3s including gp78, HRD1, TEB4, and parkin

    • This expanded interaction network likely reflects the increased complexity of protein quality control in mammals

  • Role in specialized processes:

    • Human UBE2G2 has acquired roles in processes not present in lower organisms, such as:

      • Immune regulation, as evidenced by its role in US2-mediated HLA-I degradation

      • Metabolism of specialized proteins like cytochrome P450 CYP3A4

      • Potential functions in muscle tissue, suggested by its high expression pattern

This evolutionary perspective provides insights into both the fundamental importance of UBE2G2 in eukaryotic cells and its adaptation to meet the specialized requirements of complex multicellular organisms. Comparative studies across species continue to illuminate both conserved mechanisms and novel functions of this essential E2 enzyme.

What are promising therapeutic approaches targeting UBE2G2 or its pathways?

The central role of UBE2G2 in ERAD and other cellular processes presents several promising therapeutic avenues:

  • Modulating UBE2G2 in protein misfolding diseases:

    • Small molecule inhibitors: Developing specific inhibitors of UBE2G2 could provide a targeted approach to regulate ERAD in diseases where excessive degradation of partially functional proteins occurs, such as certain cystic fibrosis mutations

    • Activators or stabilizers: Conversely, enhancing UBE2G2 activity could accelerate clearance of aggregation-prone proteins in neurodegenerative diseases

    • Approach considerations: Therapeutics would need to achieve high specificity for UBE2G2 over other E2 enzymes and carefully modulate rather than completely block activity

  • Targeting UBE2G2 in viral infections:

    • Disrupting immune evasion: Given UBE2G2's essential role in US2-mediated HLA-I downregulation , inhibitors could potentially restore immune presentation of viral antigens during cytomegalovirus infection

    • Broad-spectrum potential: This approach might extend to other viruses that exploit ERAD for immune evasion or viral protein processing

    • Delivery challenges: Targeted delivery to infected cells would be needed to minimize effects on normal ERAD function

  • UBE2G2-E3 interface targeting:

    • Selective disruption: Developing compounds that selectively disrupt specific UBE2G2-E3 interfaces (e.g., UBE2G2-TRC8) while preserving others

    • Peptidomimetics: Designing peptidomimetics based on the interacting regions of E3 ligases that selectively modify UBE2G2 activity

    • Structure-based design: Leveraging detailed structural information about the UBE2G2-E3 interface for rational drug design

  • Exploiting UBE2G2 in cancer therapy:

    • ER stress vulnerability: Cancer cells often operate under heightened ER stress and may be more vulnerable to disruption of ERAD

    • Combination approaches: Combining UBE2G2 modulation with proteasome inhibitors like bortezomib to enhance proteotoxic stress

    • Biomarker potential: UBE2G2 expression or activity levels could serve as biomarkers for sensitivity to ERAD-targeting therapies

  • Gene therapy approaches:

    • Expression modulation: AAV-mediated delivery of UBE2G2 to enhance ERAD in tissues with insufficient protein quality control

    • Engineered variants: Developing engineered UBE2G2 variants with altered specificity or activity for therapeutic applications

    • CRISPR-based approaches: Precise editing of UBE2G2 or its regulatory elements to modulate expression in specific tissues

For all these approaches, careful consideration of the essential nature of ERAD in normal cellular homeostasis is crucial, with strategies likely needing to focus on modulation rather than complete inhibition of UBE2G2 function.

What novel methodologies are emerging for studying UBE2G2 dynamics and interactions?

Cutting-edge technologies are revolutionizing our ability to study UBE2G2's dynamics and interactions with unprecedented resolution:

  • Advanced structural biology approaches:

    • Cryo-electron microscopy (cryo-EM): Enables visualization of UBE2G2 in complex with E3 ligases and substrates, potentially capturing transient intermediates in the ubiquitination cascade

    • Integrative structural biology: Combining NMR, X-ray crystallography, and cryo-EM with computational modeling to build comprehensive structural models of UBE2G2-containing complexes

    • Time-resolved structural methods: Techniques like time-resolved X-ray crystallography or T-jump NMR to capture conformational changes during catalysis

  • Single-molecule approaches:

    • Single-molecule FRET: Monitoring conformational changes in UBE2G2 during catalysis by labeling key regions with fluorescent dyes

    • Optical tweezers: Studying the mechanical aspects of ubiquitin chain formation and transfer

    • Single-molecule imaging in cells: Tracking individual UBE2G2 molecules in living cells to understand their dynamics and interactions with ER-resident partners

  • Advanced proteomics methodologies:

    • Crosslinking mass spectrometry (XL-MS): Capturing transient protein-protein interactions in the UBE2G2 interactome

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Mapping conformational changes and binding interfaces with high resolution

    • Targeted proteomics: Quantifying UBE2G2-dependent ubiquitination of specific substrates using selected reaction monitoring or parallel reaction monitoring

  • Cellular imaging innovations:

    • Super-resolution microscopy: Techniques like STORM or PALM to visualize UBE2G2 localization and dynamics at the ER membrane with nanometer precision

    • Lattice light-sheet microscopy: Enabling long-term imaging of UBE2G2 dynamics with minimal phototoxicity

    • Split fluorescent protein systems: Visualizing UBE2G2 interactions with E3 partners in real-time in living cells

  • Genetic and chemical biology tools:

    • Engineered allosteric switches: Creating conditional variants of UBE2G2 that can be controlled by light (optogenetics) or small molecules

    • Proximity labeling: BioID or APEX2 fusions to map the local environment of UBE2G2 at the ER membrane

    • Degron technologies: Rapidly controlling UBE2G2 levels using auxin-inducible or dTAG degron systems

  • Computational approaches:

    • Molecular dynamics simulations: Modeling the dynamics of UBE2G2's catalytic loops and their interactions with substrates

    • Machine learning: Predicting UBE2G2-dependent ubiquitination sites or identifying novel inhibitors

    • Systems biology modeling: Integrating UBE2G2 into comprehensive models of the ERAD pathway

These emerging methodologies promise to provide unprecedented insights into the dynamic behavior of UBE2G2, its molecular interactions, and its integration into cellular pathways, potentially revealing new opportunities for therapeutic intervention.

How might UBE2G2 research inform our understanding of broader cellular quality control mechanisms?

UBE2G2 research serves as a critical entry point for elucidating broader principles in cellular quality control and proteostasis networks:

  • Integration of quality control pathways:

    • UBE2G2's role in ERAD connects to other quality control systems including cytosolic protein quality control, autophagy, and the unfolded protein response

    • Understanding how UBE2G2 activity is coordinated with these parallel pathways can reveal principles of cellular decision-making in proteostasis

    • Research on UBE2G2 can illuminate how cells prioritize different degradation pathways and allocate resources during stress conditions

  • Selectivity in protein degradation:

    • UBE2G2's ability to assemble K48-linked polyubiquitin chains and transfer them to specific substrates raises fundamental questions about degradation selectivity

    • This research can reveal principles governing how quality control machinery distinguishes between normal proteins, those that can be refolded, and those requiring degradation

    • Insights from UBE2G2 studies may extend to other selective degradation systems throughout the cell

  • Stress adaptation mechanisms:

    • UBE2G2-dependent ERAD is crucial for cellular adaptation to ER stress

    • Understanding how UBE2G2 activity is regulated during different stress conditions provides insights into cellular adaptation strategies

    • This research informs broader questions about how cells modify their quality control thresholds during acute versus chronic stress

  • Evolution of quality control systems:

    • The conservation of UBE2G2/Ubc7 from yeast to humans, alongside the emergence of paralogous genes and expanded E3 networks, illustrates how quality control systems evolve

    • Comparative studies across species can reveal both fundamental principles and specialized adaptations in protein quality control

    • These insights contribute to our understanding of how increased complexity in multicellular organisms necessitated more sophisticated quality control mechanisms

  • Disease relevance of quality control failures:

    • UBE2G2 involvement in handling disease-associated misfolded proteins provides direct links to pathologies including:

      • Neurodegenerative diseases involving protein aggregation

      • Metabolic disorders with ER stress components

      • Immunological disorders related to antigen presentation

    • These connections highlight how quality control failures contribute to diverse disease mechanisms

  • Therapeutic implications for proteostasis disorders:

    • Understanding UBE2G2's role in quality control networks identifies potential intervention points for diseases involving proteostasis imbalance

    • Lessons from UBE2G2 research may inform strategies for other quality control components

    • The specificity of UBE2G2 for certain substrates and pathways suggests possibilities for targeted therapeutic approaches with potentially fewer side effects than global proteasome inhibition

By positioning UBE2G2 research within this broader context, scientists can leverage specific molecular insights to address fundamental questions about cellular quality control, with implications for understanding both basic biology and disease mechanisms.

Product Science Overview

Introduction to Ubiquitin-Conjugating Enzymes

Ubiquitin-conjugating enzymes, also known as E2 enzymes, play a crucial role in the ubiquitination process, which is a post-translational modification that regulates various cellular processes. These enzymes are responsible for the transfer of activated ubiquitin from the E1 ubiquitin-activating enzyme to the substrate proteins, facilitated by E3 ubiquitin ligases .

Ubiquitin-Conjugating Enzyme E2G

Ubiquitin-Conjugating Enzyme E2G, also known as UBE2G, is a specific type of E2 enzyme involved in the ubiquitination pathway. This enzyme is encoded by the UBE2G gene in humans and is essential for the regulation of protein turnover, cell cycle progression, and stress responses .

Mechanism of Action

The ubiquitination process involves a three-step enzyme cascade:

  1. Activation: The E1 enzyme activates ubiquitin in an ATP-dependent manner, forming a ubiquitin-E1 complex.
  2. Conjugation: The activated ubiquitin is transferred to the E2 enzyme (in this case, UBE2G) via a thioester bond, forming the E2-ubiquitin complex.
  3. Ligation: The E3 ubiquitin ligase facilitates the transfer of ubiquitin from the E2 enzyme to the lysine residues on the substrate protein, completing the ubiquitination process .
Biological Significance

UBE2G is involved in various cellular processes, including:

  • Protein Degradation: Ubiquitination marks proteins for degradation by the proteasome, thus regulating protein levels within the cell.
  • Cell Cycle Regulation: UBE2G plays a role in the progression of the cell cycle by targeting specific cell cycle regulators for degradation.
  • Stress Response: The enzyme is involved in the cellular response to stress by regulating the stability of stress-responsive proteins .
Disease Associations

Abnormalities in the function or expression of UBE2G have been linked to several diseases, including:

  • Cancer: Dysregulation of ubiquitination can lead to the accumulation of oncogenic proteins, contributing to tumorigenesis.
  • Neurodegenerative Diseases: Impaired ubiquitination can result in the accumulation of misfolded proteins, which is a hallmark of neurodegenerative diseases such as Parkinson’s and Alzheimer’s .
Human Recombinant UBE2G

Human recombinant UBE2G is produced using recombinant DNA technology, which involves inserting the human UBE2G gene into a suitable expression system, such as bacteria or yeast. This allows for the production of large quantities of the enzyme for research and therapeutic purposes .

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.