SUMO3 Human

Small Ubiquitin-Related Modifier 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles of SUMO3

SUMO3 modifies diverse substrates, influencing cellular pathways:

  • Nuclear processes: Regulates transcription factor activity, nucleocytoplasmic transport, and PML nuclear body assembly .

  • Stress responses: Modulates PKR activation during viral infection and protects neurons during ischemia .

  • Gene expression: SUMO2/3-specific modifications alter chromatin remodeling complexes (e.g., LSD1/CoREST/HDAC) .

Tissue-Specific Expression Patterns

Analysis of SUMO1–3 mRNA levels across human tissues reveals distinct expression profiles :

Tissue TypeSUMO1 ExpressionSUMO2 ExpressionSUMO3 Expression
LiverHighModerateLow
Reproductive OrgansModerateHighHigh
MuscleHighLowModerate

Comparative Analysis of SUMO Paralogs

SUMO paralogs exhibit functional divergence:

FeatureSUMO1SUMO2SUMO3
Sequence Identity47% vs. SUMO387% vs. SUMO387% vs. SUMO2
Chain FormationNoYesYes
Stress Response RoleActivates PKRRedundant with SUMO3Enhances PKR degradation
Tissue PrevalenceLiver, muscleUbiquitousReproductive organs

SUMO3 and SUMO2 share functional redundancy in forming poly-SUMO chains but differ in stress-specific roles. For example, SUMO3 expression accelerates PKR degradation during viral infection, unlike SUMO1 .

Non-Redundant Cellular Functions

CRISPR-Cas9 knockout studies in U2OS cells revealed SUMO3’s unique contributions :

  • Cell morphology: SUMO3 depletion disrupts cytoskeletal organization.

  • PML nuclear bodies: SUMO3 stabilizes PML body structure independently of SUMO1/2.

  • Gene regulation: SUMO3-specific substrates influence stress-responsive genes.

Isoform-Specific Splicing

Alternative splicing produces SUMO3α, an isoform with a 38-amino acid N-terminal extension. Though SUMO3α transcripts are rare (<4% in most cells), they increase under stress (e.g., heat shock) .

Pathogen Interactions

Viruses like EMCV exploit SUMO3 to downregulate PKR, suppressing antiviral defenses . SUMO3 conjugation peaks early in infection (2 hours) but declines as PKR is degraded .

Recombinant SUMO3 Applications

Recombinant SUMO3 (e.g., R&D Systems UL-762, OriGene TP720508) is widely used to study:

  • Enzyme assays: Measures SUMOylation efficiency with E1/E2 enzymes .

  • Structural studies: NMR and crystallography resolve SUMO3-Ubc9 binding interfaces .

Therapeutic Implications

SUMO3’s role in neuroprotection and viral immune evasion highlights its potential as a drug target. Inhibiting SUMO3-mediated PKR degradation could enhance antiviral responses, while promoting SUMO3 conjugation might mitigate ischemic brain damage .

Product Specs

Introduction
SUMO3, a member of the SUMO protein family, functions similarly to ubiquitin. However, unlike ubiquitin's role in protein degradation, SUMO3 participates in various cellular processes such as nuclear transport, transcriptional regulation, apoptosis, and protein stability. SUMO3 regulates amyloid beta production and plays a critical role in the development and progression of Alzheimer's disease.
Description
Produced in E. coli, this SUMO3 protein is a single, non-glycosylated polypeptide chain consisting of 112 amino acids (1-92a.a.) with a molecular mass of 12.6kDa. Note that its molecular weight on SDS-PAGE will appear higher. This SUMO3 protein is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The SUMO3 protein solution (1mg/ml) is supplied in a 20mM Tris-HCl buffer (pH 8.0) containing 0.1M NaCl, 1mM DTT, and 20% glycerol.
Stability
For optimal storage, refrigerate at 4°C if using the entire vial within 2-4 weeks. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Synonyms
SUMO-3, SMT3A, SMT3H1, SMT3 homolog 1, Small Ubiquitin-related modifier 3, Ubiquitin-like protein SMT3A.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSEEKPKEGV KTENDHINLK VAGQDGSVVQ FKIKRHTPLS KLMKAYCERQ GLSMRQIRFR FDGQPINETD TPAQLEMEDE DTIDVFQQQT GG

Q&A

What is SUMO3 and how does it differ from other SUMO paralogs?

SUMO3 is one of five human SUMO paralogs, part of the small ubiquitin-like modifier family of proteins involved in post-translational modification. Human SUMO paralogs share sequence homologies ranging from 45% to 97% . SUMO3 is synthesized as a 103 amino acid (11.5 kDa) propeptide containing an 11 amino acid C-terminal prosegment . While SUMO1 and SUMO2 are the most extensively studied paralogs, SUMO3 shares approximately 83% sequence identity with other SUMO proteins and contains the conserved ubiquitin domain and C-terminal diglycine cleavage/attachment site that are characteristic of all SUMO proteins . Unlike ubiquitination which targets proteins for degradation, sumoylation by SUMO3 participates in various cellular processes including nuclear transport, transcriptional regulation, apoptosis, and protein stability .

How does SUMO3 conjugation occur at the molecular level?

SUMO3 conjugation (sumoylation) follows a cascade similar to ubiquitination but utilizes SUMO-specific enzymes. After prosegment cleavage from the SUMO3 propeptide, the exposed C-terminal glycine forms an isopeptide bond with lysine residues on target proteins . This process requires ATP and involves three enzymatic steps: activation by E1 (SUMO-activating enzyme), transfer to E2 (SUMO-conjugating enzyme Ubc9), and finally attachment to substrates, often facilitated by E3 SUMO ligases. SUMO3 typically targets proteins containing a consensus motif ΨKxE (where Ψ is a hydrophobic residue, K is the target lysine, x is any amino acid, and E is glutamic acid), though non-consensus sumoylation also occurs .

What role does alternative splicing play in SUMO3 function?

Alternative splicing substantially contributes to SUMO3 functional diversity. Recent research has identified alternatively spliced SUMO3 transcripts that encode protein isoforms called "SUMO alphas" (SUMO3α) . These alternative transcripts change in abundance and nuclear export patterns during cellular stress. Unlike SUMO1α and SUMO2α which are non-conjugatable to protein targets, SUMO3α maintains conjugation capability but appears to target a different subset of proteins compared to canonical SUMO3 . This differential targeting suggests that alternative splicing of SUMO3 expands its functional repertoire and may represent an additional regulatory mechanism for sumoylation-dependent processes under specific cellular conditions.

How does SUMO3 expression respond to different types of cellular stress?

SUMO3 exhibits distinct response patterns to various cellular stressors. The table below summarizes key differential responses of SUMO3 to common stress conditions:

Stress ConditionSUMO3 ResponseFunctional Implication
Heat shockIncreased conjugationEnhanced protein stability and stress granule formation
Cold shockAltered splicing patternsChanges in SUMO3α levels and target specificity
Viral infection (e.g., EMCV)Enhanced conjugation to PKRModulation of antiviral responses
Oxidative stressChanges in SUMO3 distributionProtection of nuclear proteins from oxidative damage

These stress-specific responses highlight SUMO3's role in cellular adaptation mechanisms and suggest specialized functions in different stress response pathways .

How does SUMO3 modification contribute to ALS pathogenesis?

In amyotrophic lateral sclerosis (ALS) models, SUMO3 modification significantly impacts disease-related protein aggregation. Research demonstrates that SUMO3 modifies ALS-linked SOD1 mutant proteins at lysine 75 in motoneuronal cells . Compared to SUMO1, SUMO3 modification has more profound effects on SOD1 mutant proteins: it significantly increases their stability and accelerates intracellular aggregate formation . These findings suggest that SUMO3-specific modification pathways contribute to the protein aggregation processes underlying ALS pathogenesis. The differential effects between SUMO paralogs highlight the importance of studying SUMO3-specific functions in neurodegenerative disease contexts and suggest potential therapeutic targets for intervention in protein aggregation disorders .

What is the relationship between SUMO3 and antiviral responses?

SUMO3 plays a complex role in antiviral immunity through its interaction with PKR (double-stranded RNA-dependent protein kinase). Unlike SUMO1, which enhances PKR activation, SUMO3 expression reduces PKR and eIF-2α activation upon viral infection or dsRNA transfection . During viral infection with encephalomyocarditis virus (EMCV), both SUMO1 and SUMO3 conjugation to PKR increases, but with differential functional outcomes. SUMO3 expression alters PKR subcellular localization, causing concentration around the perinuclear membrane and recruitment from small speckles to nuclear dots . These findings reveal that SUMO3 may function as a regulatory mechanism to fine-tune antiviral responses, preventing excessive PKR activation that could lead to premature cell death during infection.

How do SUMO3 paralog-specific functions affect cellular responses to stress?

The paralog-specific functions of SUMO3 in stress responses reflect its unique evolutionary role. Systematic analysis using knockout cell lines and gene expression data has revealed that despite sequence similarities, SUMO3 exhibits non-redundant functions compared to other SUMO paralogs . During stress responses, SUMO3 conjugation increases substantially, but the subset of proteins modified by SUMO3 differs from those targeted by SUMO1 or SUMO2 . These paralog-specific differences extend to the regulation of various cellular processes including transcription, RNA processing, and protein quality control. Understanding these specialized functions requires comprehensive proteomic approaches that can distinguish between SUMO paralog-specific modification events and their downstream consequences in different cellular contexts .

What are the optimal approaches for detecting SUMO3-modified proteins?

Detecting SUMO3-modified proteins requires specialized techniques due to the often transient and substoichiometric nature of sumoylation. For immunodetection, monoclonal antibodies specifically recognizing human SUMO3 (such as clone 401513R) can be used for immunofluorescence, Western blotting, and immunoprecipitation applications . For localization studies, immunofluorescence with appropriate secondary antibodies can visualize SUMO3 in fixed cells, with nuclear counterstaining revealing its predominant nuclear localization .

For proteomic identification of SUMO3 targets, approaches include:

  • Expression of tagged SUMO3 (His, FLAG, or HA) followed by affinity purification

  • SUMO-remnant antibodies recognizing the diglycine motif left after trypsin digestion

  • Proximity-based labeling methods using SUMO3 fused to biotin ligases

  • Stable isotope labeling with amino acids in cell culture (SILAC) combined with mass spectrometry

These approaches should be complemented with site-directed mutagenesis of potential SUMO3 attachment sites (lysine residues) to confirm direct modification of candidate proteins .

How can researchers utilize SUMO3 fusion technology in protein expression systems?

SUMO3 fusion technology offers significant advantages for recombinant protein expression, particularly for difficult-to-express proteins. The SUMOpro-3 Gene Fusion system enables convenient directional cloning of a gene of interest (GOI) in frame with the SUMO3 fusion tag . This approach dramatically enhances protein expression and promotes solubility and correct folding of the target protein .

Implementation requires:

  • Design of appropriate primers incorporating BsaI restriction sites for directional cloning

  • PCR amplification of the target gene with these primers

  • Cloning into pE-SUMO3 vector (available with ampicillin or kanamycin resistance markers)

  • Expression in BL21(DE3) or related E. coli strains

  • Purification followed by SUMO Protease 2 cleavage to remove the SUMO3 tag

The system's key advantage is that SUMO Protease 2 has no known instances of cleaving within the protein of interest, and cleavage yields the native protein with the desired N-terminus (except when proline is the first residue) . This methodology is particularly valuable for expressing human proteins that typically show poor solubility in bacterial expression systems.

What controls and validation steps are essential in SUMO3 research?

Rigorous SUMO3 research requires multiple controls and validation steps:

  • Paralog specificity controls: Include SUMO1, SUMO2, and SUMO3 in parallel experiments to distinguish paralog-specific effects from general sumoylation consequences .

  • SUMO3 mutant controls: Use conjugation-deficient SUMO3 mutants (e.g., diglycine motif mutants) to distinguish between conjugation-dependent and protein-protein interaction effects .

  • Site-specific validation: For putative SUMO3 targets, create lysine-to-arginine mutants at predicted conjugation sites and verify loss of sumoylation .

  • Stress condition controls: Include appropriate positive controls for stress responses when studying stress-induced SUMO3 modifications .

  • Antibody validation: Confirm antibody specificity using SUMO3 knockout cell lines or siRNA-mediated knockdown to prevent cross-reactivity with other SUMO paralogs .

  • Functional validation: After identifying SUMO3-modified proteins, perform functional assays to determine the biological significance of the modification beyond mere detection .

These controls help ensure that observed effects are specifically attributable to SUMO3 and not to other SUMO paralogs or non-specific experimental artifacts.

What are the functional consequences of SUMO3α compared to canonical SUMO3?

Alternative splicing produces SUMO3α, which differs functionally from canonical SUMO3. Unlike SUMO1α and SUMO2α which are non-conjugatable, SUMO3α retains the ability to conjugate to target proteins but appears to modify a different subset of proteins compared to canonical SUMO3 . This suggests that SUMO3α may regulate specific cellular processes distinct from those controlled by canonical SUMO3. The differential cellular distribution of SUMO3α compared to canonical SUMO3 further supports this functional specialization . Additionally, the expression and nuclear export of SUMO3α are affected by cellular stress in a stress-specific and cell-type dependent manner, suggesting context-dependent roles . Understanding these differences is critical for comprehensive analysis of SUMO3 functions in various physiological and pathological contexts.

What evolutionary insights can be gained from studying SUMO3-specific functions?

Evolutionary analysis of SUMO paralogs provides insights into their specialized functions. The existence of multiple SUMO paralogs in mammals compared to single SUMO proteins in lower eukaryotes suggests that diversification of the SUMO system allowed for more complex regulatory networks during evolution . SUMO3's conservation across species indicates its fundamental importance, while species-specific differences in SUMO3 regulation and target profiles reflect evolutionary adaptation to different environmental challenges . The conservation of specific lysine residues as SUMO3 attachment sites across species (as seen in proteins like SOD1 and PKR) further supports the biological significance of these modification events . These evolutionary insights help researchers understand why specific SUMO3 functions cannot be compensated by other paralogs and highlight the evolutionary pressure that maintained these specialized functions.

What are the key technical limitations in current SUMO3 research?

Despite significant advances, SUMO3 research faces several technical challenges:

  • Paralog discrimination: The high sequence similarity between SUMO2 and SUMO3 (approximately 97%) makes it difficult to develop truly paralog-specific antibodies and detection methods .

  • Transient modifications: SUMO3 modifications are often dynamic and substoichiometric, making their detection challenging without enrichment strategies .

  • Context-dependent effects: SUMO3 functions vary significantly depending on cell type, stress conditions, and other contextual factors, necessitating complex experimental designs to capture this variability .

  • Alternative transcript detection: Current methods may underestimate the complexity of SUMO3 alternative splicing, potentially missing important regulatory mechanisms .

  • Physiological relevance: Distinguishing between physiologically relevant SUMO3 modifications and artificial effects due to overexpression systems remains challenging .

Addressing these limitations requires developing more sensitive and specific tools for detecting endogenous SUMO3 modifications and their dynamic changes under various physiological conditions.

What emerging technologies are advancing SUMO3 research?

Several emerging technologies are poised to transform SUMO3 research:

  • CRISPR-based approaches: CRISPR/Cas9 genome editing enables generation of paralog-specific knockouts and endogenous tagging of SUMO3, allowing more physiologically relevant studies .

  • Advanced proteomics: Quantitative multiplexed proteomics combined with specific enrichment strategies can now identify SUMO3-modified proteomes with unprecedented depth and accuracy .

  • Single-cell technologies: Single-cell transcriptomics and proteomics are beginning to reveal cell-to-cell variability in SUMO3 expression and targets, providing insights into heterogeneous cellular responses .

  • Live-cell imaging: New fluorescent sensors for SUMO3 modification enable real-time visualization of dynamic sumoylation events in living cells .

  • Structural biology advances: Cryo-electron microscopy and integrative structural biology approaches are providing detailed insights into how SUMO3 modifies target protein structure and function .

These technologies will facilitate more comprehensive understanding of SUMO3 biology and potentially reveal new therapeutic targets in SUMO3-related disorders.

How might SUMO3-targeted therapeutic approaches develop in the future?

The growing understanding of SUMO3 in disease contexts suggests several potential therapeutic directions:

  • Neurodegenerative diseases: Given SUMO3's role in accelerating SOD1 aggregation in ALS, inhibitors specifically targeting SUMO3 conjugation to SOD1 could represent a therapeutic strategy for slowing disease progression .

  • Viral infections: Modulators of SUMO3's effects on PKR activation could potentially enhance antiviral responses in a more controlled manner than direct PKR activators, reducing off-target effects .

  • Cancer therapy: As SUMO3 regulates multiple oncogenic and tumor suppressor proteins, cancer-specific SUMO3 inhibitors might offer therapeutic windows not available with general sumoylation inhibitors .

  • Stress response modulation: Compounds targeting the alternative splicing of SUMO3 could potentially modulate cellular stress responses in conditions like ischemia-reperfusion injury .

Product Science Overview

Introduction

Small Ubiquitin-Related Modifier 3 (SUMO3) is a member of the SUMO family of proteins, which are ubiquitin-like proteins involved in post-translational modification of other proteins. SUMOylation, the process of attaching SUMO proteins to target proteins, plays a crucial role in various cellular processes, including nuclear transport, transcriptional regulation, apoptosis, protein stability, response to stress, and cell cycle progression .

Structure and Function

SUMO3, like other SUMO proteins, is covalently attached to target proteins via an isopeptide bond between the C-terminal glycine residue of SUMO3 and an acceptor lysine on the target protein . This modification can alter the localization, stability, and interaction partners of the target protein, thereby influencing its function .

SUMO3 is highly similar to SUMO2, with which it shares a high degree of sequence identity . Both SUMO2 and SUMO3 are distinct from SUMO1, another member of the SUMO family . SUMO3 is involved in various cellular processes, including nuclear transport, DNA replication and repair, mitosis, and signal transduction .

SUMOylation Process

The SUMOylation process involves a cascade of enzymatic reactions similar to ubiquitination. It begins with the activation of SUMO3 by the E1 enzyme complex (SAE1-SAE2), followed by its conjugation to the E2 enzyme (UBE2I). The final step involves the transfer of SUMO3 to the target protein, which can be facilitated by E3 ligases such as PIAS1-4, RANBP2, or CBX4 .

Biological Significance

SUMOylation is a reversible process, and the removal of SUMO3 from target proteins is mediated by deSUMOylating enzymes . This dynamic modification allows cells to rapidly respond to various stimuli and stress conditions. SUMO3 modification has been shown to regulate protein stability, nuclear-cytosolic transport, and transcriptional activity .

Applications of Human Recombinant SUMO3

Human recombinant SUMO3 is widely used in research to study the SUMOylation process and its effects on target proteins. It is also used to investigate the role of SUMO3 in various cellular processes and diseases. Recombinant SUMO3 can be produced in large quantities using bacterial expression systems, making it a valuable tool for biochemical and structural studies.

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.