SPOP Human

Speckle-Type POZ Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to SPOP Human

Speckle-type POZ protein (SPOP), encoded by the SPOP gene (chromosome 17), is a critical adaptor protein for the CUL3-RBX1 E3 ubiquitin ligase complex. It facilitates substrate ubiquitination and degradation, regulating diverse cellular processes such as DNA repair, transcriptional repression, and tumorigenesis. SPOP’s structure includes:

  • MATH domain (residues 31–164): Binds substrates via a conserved SPOP-binding consensus (SBC) motif (Φ-π-S-S/T-S/T) .

  • BTB/POZ domain (residues 184–297): Mediates dimerization and interaction with CUL3 .

  • BACK domain (residues 300–359): Enhances oligomerization and substrate avidity .

SPOP’s primary role involves targeting oncogenic proteins for degradation, acting as a tumor suppressor in prostate, kidney, and endometrial cancers . Mutations in its MATH domain disrupt substrate binding, leading to oncogenic effects .

Role in DNA Repair and Replication

SPOP regulates DNA-protein crosslink repair by removing topoisomerase 2A from cleavage complexes . In replication, SPOP promotes K27-linked polyubiquitination of Geminin, preventing excessive origin firing and re-replication .

ProcessMechanismClinical ImpactReferences
DNA RepairRemoves topoisomerase 2A from DNA-protein crosslinksCritical for genome stability
DNA ReplicationUbiquitinates Geminin (K100/K127) to block Cdt1-MCM complex formationPrevents replication catastrophe

Mutations in SPOP (e.g., Y87C, F133S) disrupt Geminin ubiquitination, causing replication stress and sensitivity to ATR inhibitors .

Cancer-Related Functions

SPOP mutations occur in ~15% of prostate cancers and ~13% of endometrial cancers, acting as a tumor suppressor .

Prostate Cancer

  • Wild-type SPOP: Degrades oncogenic proteins (e.g., ERG, SRC3) .

  • Mutant SPOP: Fails to degrade ZMYND11, enhancing androgen receptor (AR) signaling and promoting castration-resistant growth .

Kidney Cancer

  • SPOP overexpression: Targets tumor suppressors (e.g., PTEN, SETD2) for degradation, promoting tumorigenesis .

Regulation of Adipogenesis and Thermogenesis

In mice, SPOP knockout impairs adipogenesis and upregulates thermogenic markers (e.g., UCP1) :

PhenotypeMechanismOutcomeReferences
Impaired AdipogenesisReduces PPARγ-driven differentiation in preadipocytesResistance to obesity
Enhanced ThermogenesisUpregulates UCP1, COX6C2, Ndufb2 in brown/beige adipose tissueIncreased energy expenditure

Regulation of Fetal Hemoglobin (HbF)

SPOP represses γ-globin via CUL3-dependent ubiquitination. Inhibition of SPOP increases HbF production, offering therapeutic potential for β-hemoglobinopathies :

Cell ModelEffect of SPOP DepletionMechanismReferences
HUDEP-2 Erythroid Cells↑ γ-globin mRNA/protein; ↑ HbF+ cell populationCUL3-dependent repression
Primary CD34+ Cells↑ γ-globin levels; minimal impact on erythroid maturationIndependent of BCL11A/LRF

Therapeutic Implications

StrategyTargetPotential ApplicationReferences
SPOP InhibitorsERG-fused prostate cancerSensitizes to high-dose androgen therapy
ATR InhibitorsSPOP-mutant cancersInduces replication catastrophe
SPOP Depletionβ-hemoglobinopathiesReactivates HbF production

Substrates and Pathways

SubstrateFunctionSPOP InteractionReferences
GemininRegulates DNA replication origin licensingK27-linked polyubiquitination
IRF2BP2Suppresses HCC cell proliferation/migrationTargets for degradation via MATH domain
ZMYND11Represses ERG-dependent oncogenesisDegradation via wild-type SPOP
γ-globinFetal hemoglobin synthesisCUL3-dependent repression

Product Specs

Introduction
Speckle-type POZ protein (SPOP) is a member of the Tdpoz family. It possesses a MATH (Meprin and TRAF homology) domain at the N-terminus and a BTB/POZ domain at the C-terminus. SPOP can hinder IPF1/PDX1's ability to activate target promoters, potentially by assembling a repressor complex. It plays a role in ubiquitinylation and protein degradation through its interaction with CUL-3.
Description
Recombinant human SPOP, with a 20 amino acid His tag at its N-terminus, is produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 394 amino acids (residues 1-374) and has a molecular weight of 44.3 kDa. The purification of SPOP is achieved using proprietary chromatographic techniques.
Physical Appearance
SPOP is provided as a sterile, colorless, and filtered solution.
Formulation
The SPOP solution has a concentration of 0.5 mg/ml and consists of 20mM Tris-HCl buffer (pH 8.0), 5mM DTT, 50% glycerol, 0.2M NaCl, and 2mM EDTA.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms

Speckle-type POZ protein, HIB homolog 1, Roadkill homolog 1, SPOP, TEF2.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MSRVPSPPPP AEMSSGPVAE SWCYTQIKVV KFSYMWTINN FSFCREEMGE VIKSSTFSSG ANDKLKWCLR VNPKGLDEES KDYLSLYLLL VSCPKSEVRA KFKFSILNAK GEETKAMESQ RAYRFVQGKD WGFKKFIRRD FLLDEANGLL PDDKLTLFCE
VSVVQDSVNI SGQNTMNMVK VPECRLADEL GGLWENSRFT DCCLCVAGQE FQAHKAILAA RSPVFSAMFE HEMEESKKNR VEINDVEPEV FKEMMCFIYT GKAPNLDKMA DDLLAAADKY ALERLKVMCE DALCSNLSVE NAAEILILAD LHSADQLKTQ AVDFINYHAS DVLETSGWKS MVVSHPHLVA EAYRSLASAQ CPFLGPPRKR LKQS.

Q&A

What is SPOP and what are its primary cellular functions?

SPOP is a substrate-binding adaptor of the CUL3-RING E3 ubiquitin ligase complex that mediates the ubiquitination of target proteins, typically leading to their proteasomal degradation . The protein contains two primary functional domains: the MATH domain in its N-terminus for substrate recognition and the BTB domain in its C-terminus that scaffolds with the CUL3-RING complex .

SPOP regulates numerous cellular processes by targeting various substrate proteins. Notable substrates include GLI2, PD-L1, NANOG, TRIM24, CYCLIN E1, and c-MYC, many of which are oncoproteins . Additionally, SPOP plays essential roles in DNA repair, particularly in resolving DNA-protein crosslinks by removing topoisomerase 2A from DNA cleavage complexes .

Research methodology: To study SPOP's cellular functions, researchers typically employ gene knockdown/knockout approaches using siRNA/shRNA or CRISPR-Cas9, followed by functional assays examining effects on ubiquitination, protein stability, cell proliferation, and DNA repair capacity.

What are the common mutations in SPOP and their prevalence in human cancers?

SPOP mutations are particularly significant in prostate cancer, where they represent one of the most common point mutations. Studies have identified SPOP mutations in:

  • 6-13% of primary prostate adenocarcinomas

  • 14.5% of metastatic prostate cancers

  • 4.6-14.4% of prostate cancer patients across different ethnic backgrounds

Notably, SPOP mutations have been observed in high-grade prostatic intraepithelial neoplasia (HG-PIN) adjacent to invasive adenocarcinoma, suggesting they are early events in prostate tumorigenesis . These mutations typically occur in the MATH domain, potentially altering substrate recognition .

Research methodology: Mutation detection typically involves targeted gene sequencing or whole-exome/genome sequencing of tumor samples compared to matched normal tissues. For functional validation, site-directed mutagenesis is used to generate mutant SPOP constructs for cellular assays.

How do experimental models for SPOP research differ, and what are their advantages?

Several experimental models have been developed to study SPOP function:

Cell line models:

  • Overexpression systems: Useful for studying gain-of-function effects

  • CRISPR-Cas9 knockout/knockin: Enables precise genetic manipulation to study loss-of-function or mutation effects

  • WRL68 human normal hepatocytes have been used to study SPOP's effects on gene expression profiles and alternative splicing

Mouse models:

  • Conditional expression systems such as R26-F133V/+ mice allow for tissue-specific induction of SPOP mutations

  • Primary murine prostate cell (MPC) lines can be derived from these mice for in vitro studies

Zebrafish models:

  • In vivo mRNA rescue experiments enable studying evolutionary conservation of SPOP function

Research methodology: Model selection depends on research questions. For basic molecular mechanisms, cell lines offer simplicity and throughput. For physiological relevance and tumor microenvironment studies, mouse models are preferred. Combined approaches provide complementary insights.

How do SPOP mutations affect substrate preference and functional outcomes?

Recent research challenges the simple view that SPOP mutations merely disrupt substrate binding. A study investigating the HCC-derived mutant SPOP-M35L revealed it actually increases affinity to the tumor suppressor IRF2BP2 compared to wild-type SPOP . This mutation appears to reprogram SPOP from a tumor suppressor to an oncoprotein, promoting hepatocellular carcinoma (HCC) cell proliferation and metastasis .

This example illustrates that SPOP mutations can have context-dependent effects:

Research methodology: To investigate mutation effects on substrate preference, researchers employ:

  • Co-immunoprecipitation assays comparing wild-type and mutant SPOP binding to various substrates

  • Ubiquitination assays to measure functional consequences on substrate degradation

  • Structural biology approaches (X-ray crystallography, cryo-EM) to understand molecular interactions

  • Cellular assays measuring proliferation, migration, and other cancer-related phenotypes

What is SPOP's role in genomic instability and DNA repair mechanisms?

SPOP mutations contribute to genomic instability in prostate cancer by altering DNA repair processes . SPOP is essential for the repair of DNA-protein crosslinks, specifically by removing topoisomerase 2A from the topoisomerase2A-DNA cleavage complex formed during repair .

The exact mechanism involves:

  • Recognition of topoisomerase 2A by SPOP's MATH domain

  • Recruitment of the CUL3-RING E3 ligase complex via the BTB domain

  • Ubiquitination of topoisomerase 2A, facilitating its removal from DNA

  • Resolution of the DNA-protein crosslink to maintain genomic stability

When SPOP is mutated, this process is disrupted, potentially leading to:

  • Persistence of DNA-protein crosslinks

  • Increased DNA damage

  • Genomic instability

  • Accelerated tumorigenesis

Research methodology: To study SPOP's role in DNA repair, researchers use:

  • DNA damage assays (comet assay, γ-H2AX foci)

  • DNA-protein crosslink detection methods

  • Time-course analysis of repair kinetics

  • Genomic instability measurements (micronuclei, chromosomal aberrations)

How does SPOP regulate gene expression profiles and alternative splicing?

SPOP has emerged as a significant regulator of gene expression and alternative splicing, particularly relevant in hepatocellular carcinoma (HCC). RNA sequencing of SPOP-overexpressing WRL68 human normal hepatocytes identified 3,838 differentially expressed genes (DEGs), including 1,522 upregulated and 2,316 downregulated genes .

SPOP's regulation of gene expression involves:

  • Direct ubiquitination of transcription factors and epigenetic regulators

  • Modulation of chromatin structure through interaction with histone-associated proteins

  • Regulation of alternative splicing events

A comprehensive assessment revealed SPOP-regulated alternative splicing events are involved in pathways associated with:

  • Cellular processes

  • Metabolism

  • Environmental information processing

  • Organismal systems

Research methodology: To investigate SPOP's effects on gene expression and splicing:

  • RNA sequencing with differential expression analysis

  • Alternative splicing analysis using computational tools like rMATS

  • Validation of key targets using qRT-PCR

  • Functional pathway enrichment analysis using GO terms and KEGG pathways

What is the current understanding of SPOP's dual role as both tumor suppressor and oncogene?

SPOP exhibits context-dependent functions that can be either tumor-suppressive or oncogenic, challenging the conventional view of cancer genes as exclusively one or the other.

Tumor suppressor evidence:

  • Wild-type SPOP suppresses HCC cell proliferation and metastasis

  • Many SPOP substrates are oncoproteins (GLI2, PD-L1, NANOG, TRIM24, CYCLIN E1, c-MYC)

  • Loss-of-function mutations are common in certain cancers

Oncogenic evidence:

  • SPOP overexpression facilitated cell proliferation in human normal hepatocytes

  • The HCC-derived SPOP-M35L mutant promotes cancer cell proliferation and metastasis

  • SPOP may potentially exhibit tumor-promoting effects in certain contexts

This duality appears to depend on:

  • Tissue-specific expression of SPOP substrates

  • Mutation-specific effects on substrate preference

  • The balance of pro- and anti-tumorigenic substrates in a given cellular context

Research methodology: To investigate this dual role, researchers should:

  • Perform context-specific knockdown/overexpression experiments

  • Compare wild-type and mutant SPOP effects across multiple cancer types

  • Identify tissue-specific substrates through proteomics approaches

  • Use in vivo models to validate in vitro findings

What are promising therapeutic approaches targeting SPOP or its pathway?

Given SPOP's role in cancer development, several therapeutic strategies warrant investigation:

  • Substrate-stabilizing compounds:

    • Design molecules that prevent SPOP-substrate interaction

    • Particularly valuable for contexts where SPOP mutations drive oncogenesis

  • PROTAC (Proteolysis Targeting Chimera) approach:

    • Utilize SPOP's substrate recognition mechanism to target oncoproteins for degradation

    • Design bifunctional molecules that bring together SPOP and specific cancer-promoting proteins

  • Synthetic lethality strategies:

    • Identify vulnerabilities created by SPOP mutations

    • Target compensatory pathways activated in SPOP-mutant cancers

Research methodology: Drug development approaches include:

  • Structure-based drug design targeting the MATH or BTB domains

  • High-throughput screening of compound libraries

  • PROTAC design and optimization

  • Functional genomics (CRISPR screens) to identify synthetic lethal interactions

How can genomic and proteomic approaches advance SPOP research?

Integrated multi-omics approaches offer promising avenues for SPOP research:

Genomic approaches:

  • Comprehensive mutation profiling across cancer types

  • CRISPR-Cas9 screens to identify synthetic lethal interactions

  • ChIP-seq to identify genomic binding sites of SPOP-regulated transcription factors

Proteomic approaches:

  • Global ubiquitinome analysis to identify novel SPOP substrates

  • Protein interaction network mapping using BioID or APEX proximity labeling

  • Phospho-proteomics to understand signaling pathways affected by SPOP

Integrated analysis:

  • Correlation of genomic alterations with proteomic changes

  • Multi-omics data integration to build predictive models

  • Single-cell approaches to understand heterogeneity in SPOP function

Research methodology: Integration of these approaches requires advanced computational methods and careful experimental design with appropriate controls and validation strategies.

Product Science Overview

Introduction

Speckle-Type POZ Protein (SPOP) is a protein encoded by the SPOP gene in humans . It is a substrate adaptor of the Cullin3 (CUL3)-based E3 ubiquitin ligase complex, playing a crucial role in the ubiquitination and degradation of various substrate proteins . SPOP is involved in several cellular processes, including transcriptional repression, cell proliferation, migration, and apoptosis .

Structure and Function

SPOP contains a typical POZ/BTB domain at the N-terminal and a MATH/TRAF domain at the C-terminal . The POZ/BTB domain binds to the ubiquitin ligase CUL3, while the MATH/TRAF domain binds to specific substrates . This dual-domain structure allows SPOP to act as a substrate adaptor, recognizing and recruiting substrate proteins for ubiquitination and subsequent degradation .

Role in Cancer

SPOP has been extensively studied for its role in various cancers. It functions as a tumor suppressor in several types of cancer, including prostate cancer and renal cell carcinoma (RCC) . In prostate cancer, SPOP promotes the degradation of bromodomain and extraterminal (BET) proteins, impacting the effectiveness of BET inhibitors . Additionally, SPOP regulates androgen receptor (AR) signaling, with down-expression leading to the activation of AR signaling and exerting oncogenic effects .

In RCC, SPOP is overexpressed in cancer tissues compared to adjacent normal tissues . Overexpression of SPOP inhibits cell proliferation, migration, and invasion while increasing cell apoptosis . These findings suggest that SPOP could be a potential tumor inhibitor in RCC .

Clinical Significance

SPOP has potential as a biomarker for cancer diagnosis and prognostic stratification . Higher expression of SPOP is associated with earlier clinical stages, better differentiation, and improved overall survival in various cancers . Its role in promoting ubiquitination-mediated degradation of programmed death ligand 1 (PD-L1) also highlights its importance in cancer immune surveillance .

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 2025 TheBiotek. All Rights Reserved.