TRIM33 regulates diverse biological pathways, including transcription, immune response, and cancer progression.
TRIM33 modulates gene expression by interacting with transcription factors and chromatin modifiers:
TGF-β/Smad Signaling: Binds SMAD2/3 and SMAD4, influencing nuclear exclusion or degradation of SMAD4, thereby repressing TGF-β target genes .
Nodal Signaling: Recruits to PML nuclear bodies in embryonic stem cells, regulating Lefty1/2 expression essential for mesendoderm differentiation .
AR Signaling: Stabilizes androgen receptor (AR) protein in prostate cancer, enhancing transcription of androgen-responsive genes .
TRIM33 is critical for innate immunity:
NLRP3 Inflammasome Activation: Ubiquitinates DHX33 (K63-linked), facilitating its interaction with NLRP3 and subsequent IL-1β/IL-18 production in response to cytosolic RNA .
Dendritic Cell (DC) Homeostasis: Regulates Irf8 (cDC1 differentiation) and Bcl2l11 (apoptosis suppression), maintaining DC survival and lineage specification .
Immune Function | Mechanism | Outcome |
---|---|---|
Cytosolic RNA sensing | DHX33 ubiquitination → NLRP3 activation | IL-1β/IL-18 secretion |
DC differentiation | Irf8 transcription → cDC1 development | Enhanced T-cell priming |
DC survival | Bcl2l11 repression → apoptosis prevention | Maintained DC homeostasis |
TRIM33 exhibits context-dependent roles in malignancy:
TRIM33 loss in multiple myeloma patients leads to:
Increased Chromosomal Aberrations: Median 41.5 structural variants vs. 27 in TRIM33-intact patients .
Impaired Mitotic Checkpoints: Downregulation of spindle assembly and G2M checkpoint pathways .
Conditional Trim33 deletion in mice causes:
Reduced DC Subsets: cDC1, cDC2, and pDC populations decline .
Impaired Immune Responses: Defective T-cell priming and antiviral defense .
TRIM33 knockdown reduces AR protein stability, while overexpression enhances:
Targeting TRIM33 in Prostate Cancer: Inhibitors to disrupt AR-TRIM33 interactions may mitigate AR-dependent growth .
Stabilizing TRIM33 in Myeloma: Restoring TRIM33 expression could enhance genomic stability and survival .
Immune Modulation: Leveraging TRIM33’s role in NLRP3 inflammasome activation for antiviral or anticancer therapies .
TRIM33 (Tripartite Motif Containing 33) is a member of the TRIM family of proteins, characterized by a tripartite motif consisting of RING, B-box, and coiled-coil domains. TRIM33 (also known as TIF1γ) is one of four TRIM family proteins—alongside TRIM24 (TIF1α), TRIM28 (TIF1β), and TRIM66—that contain C-terminal plant homeodomain (PHD) and bromodomain (BRD) modules. These specialized domains enable TRIM33 to recognize and bind to specific histone modifications: the PHD domain binds methylated lysine residues, while the bromodomain recognizes acetylated lysine residues . As a multifunctional protein, TRIM33 operates as both a transcriptional cofactor that regulates gene expression and an E3 ubiquitin ligase that catalyzes the transfer of ubiquitin to substrate proteins.
Two primary isoforms of TRIM33 have been characterized: TRIM33α and TRIM33β. The key structural difference between these isoforms relates to the orientation of the N1039 residue in their bromodomains. This residue is equivalent to N140 in BRD4(1) and is generally conserved across most bromodomains. In TRIM33β, this residue is positioned to coordinate with acetylated lysine (KAc) residues on histone tails, while in TRIM33α, this coordination does not occur due to the different residue orientation . Despite this structural distinction, biophysical studies using isothermal titration calorimetry (ITC) have shown that both isoforms display similar binding affinities for histone H3 peptides with specific modifications, particularly H3 1-27K18Ac. Both isoforms also show preferential binding to dual-modified H3 1-27K9Me3K18Ac peptides, indicating that their PHD and BRD domains contribute additively to binding affinity .
In research settings, several methodologies are employed to detect and quantify TRIM33 expression:
For protein-level detection:
Western blotting using specific anti-TRIM33 antibodies (e.g., Bethyl Laboratories A301-060A)
Immunoprecipitation followed by mass spectrometry for protein complex analysis
Immunohistochemistry or immunofluorescence for tissue or cellular localization
For mRNA-level detection:
Real-time PCR (RT-PCR) to measure TRIM33 transcript levels, with knockdown efficiency often reported as >60% reduction in mRNA levels
RNA sequencing (RNA-seq) for genome-wide expression analysis
Northern blotting for specific transcript detection
For experimental manipulation:
shRNA-mediated knockdown systems have been established in various cell types including THP-1-derived macrophages
siRNA approaches have shown efficacy in human primary monocyte-derived macrophages (hPMDM)
CRISPR-Cas9 genome editing for creating knockout models or tagged endogenous TRIM33
The selection of appropriate detection methods depends on the research question, with protein-level analysis being crucial for functional studies and mRNA analysis for transcriptional regulation studies.
TRIM33 functions as an E3 ubiquitin ligase through its N-terminal RING domain, which is characteristic of many E3 ligases. The mechanistic process involves:
Substrate recognition and binding: TRIM33 directly binds specific substrate proteins, such as the cytosolic double-stranded RNA sensor DHX33 .
Coordination with E2 enzymes: The RING domain recruits ubiquitin-loaded E2 enzymes to facilitate ubiquitin transfer.
Site-specific ubiquitination: TRIM33 catalyzes the attachment of ubiquitin to specific lysine residues on substrate proteins. For example, TRIM33 induces lysine 63 (K63)-specific ubiquitination of DHX33 at lysine 218 upon dsRNA stimulation .
Functional modification: Unlike K48-linked ubiquitination that typically targets proteins for proteasomal degradation, the K63-linked ubiquitination catalyzed by TRIM33 often modifies protein function or interactions rather than triggering degradation.
Downstream signaling: In the case of DHX33, TRIM33-mediated ubiquitination is essential for the formation of the DHX33-NLRP3 complex and subsequent inflammasome activation, leading to caspase-1 activation and production of pro-inflammatory cytokines IL-1β and IL-18 .
Experimental approaches to study this activity include in vitro ubiquitination assays, site-directed mutagenesis to generate lysine-to-arginine mutants of substrate proteins, and functional readouts such as measuring inflammasome activation following dsRNA stimulation .
TRIM33 functions as a versatile transcriptional regulator through multiple mechanisms:
Genomic binding pattern: ChIP-seq analysis has revealed that TRIM33 predominantly binds regions within 2kb of transcription start sites (TSS), with enrichment directly over the TSS , positioning it to influence gene expression initiation.
Context-dependent activation or repression: TRIM33 can both activate and repress transcription depending on the genomic context and cell type:
Transcription factor interaction: TRIM33 interacts with specific transcription factors to modulate their activity. In B cell leukemia, TRIM33 is recruited to enhancers by the transcription factor PU.1 but antagonizes PU.1's function at these sites .
Epigenetic recognition: Through its PHD and bromodomain modules, TRIM33 recognizes specific histone modifications, particularly H3K9Me3 and H3K18Ac , allowing it to "read" the epigenetic code and respond accordingly.
Enhancer regulation: TRIM33 can associate with specific enhancers to modulate gene expression, as demonstrated by its binding to an enhancer upstream of the Bim gene in B lymphoblastic leukemia cells .
Repression of repetitive elements: TRIM33 binds and silences specific classes of endogenous retroelements, particularly RLTR10B elements, showing nearly seven-fold enrichment at these sites .
This multifaceted regulatory capacity allows TRIM33 to coordinate complex transcriptional programs in various cellular contexts.
TRIM33 interacts with specific histone modifications through its PHD and bromodomain (BRD) modules. Detailed biophysical studies have characterized these interactions:
Histone Peptide | TRIM33α Binding Affinity (Kd) | TRIM33β Binding Affinity (Kd) | Primary Domain Interaction |
---|---|---|---|
H3 1-27K9Me3 | Moderate affinity | Moderate affinity | PHD domain |
H3 1-27K18Ac | Moderate affinity | Moderate affinity | Bromodomain |
H3 1-27K9Me3K18Ac | Higher affinity | Higher affinity | PHD + Bromodomain (additive) |
The dual-modified histone peptide H3 1-27K9Me3K18Ac shows the strongest interaction with both TRIM33 isoforms, demonstrating that the PHD and BRD domains contribute additively to binding affinity . This suggests that TRIM33 preferentially recognizes nucleosomes carrying both methylation at H3K9 and acetylation at H3K18, potentially allowing it to integrate multiple epigenetic signals.
Despite structural differences in their bromodomains, particularly in the orientation of the N1039 residue, both TRIM33α and TRIM33β display similar binding affinities for these histone modifications as measured by isothermal titration calorimetry (ITC) . This indicates that the functional capacity to recognize specific histone marks is preserved across isoforms, though the structural basis for this recognition may differ.
Researchers have leveraged this understanding to develop AlphaScreen assays for the TRIM33 PHD-BRD cassettes, enabling high-throughput screening to identify novel ligands that may modulate TRIM33's interaction with modified histones .
TRIM33 plays a critical role in preventing apoptosis in B lymphoblastic leukemia through a highly specific regulatory mechanism:
Lineage dependency: TRIM33 has been identified as a lineage dependency in B cell neoplasms, meaning these cancer cells rely on TRIM33 for survival .
Enhancer-specific regulation: Unlike most transcription factors that bind thousands of genomic sites, TRIM33 performs its essential anti-apoptotic function by associating with a single critical enhancer. ChIP-seq analysis revealed that TRIM33 preferentially binds to two lineage-specific enhancers containing an exceptionally high density of motifs recognized by the PU.1 transcription factor .
Antagonism of PU.1 function: Although TRIM33 is recruited to these enhancers by PU.1, it acts to antagonize PU.1's transcriptional activity. One of these PU.1/TRIM33 co-occupied enhancers is located upstream of the pro-apoptotic gene Bim (encoded by Bcl2l11) .
Suppression of pro-apoptotic signaling: By binding to the Bim enhancer, TRIM33 interferes with enhancer-mediated Bim activation, thereby preventing the expression of this key pro-apoptotic factor and subsequent apoptosis in leukemia cells .
Functional specificity: The critical nature of this single regulatory element is demonstrated by the finding that deleting this enhancer renders TRIM33 dispensable for leukemia cell survival .
This mechanism represents an unusual case in transcriptional regulation, where a cofactor performs an essential function by regulating a single genetic element rather than orchestrating broad transcriptional programs across thousands of genomic sites.
TRIM33 functions as an essential component in cytosolic RNA-induced inflammasome activation, a critical process in innate immune responses:
RNA sensing pathway: When cytosolic double-stranded RNA (dsRNA) is detected, such as during viral infection, it triggers a cascade of events leading to inflammasome activation .
DHX33 ubiquitination: TRIM33 directly binds the cytosolic dsRNA sensor DHX33 and catalyzes its ubiquitination via lysine 218 upon dsRNA stimulation .
K63-specific ubiquitination: TRIM33 induces lysine 63 (K63)-specific ubiquitination of DHX33, which does not target it for degradation but rather modifies its functional interactions .
DHX33-NLRP3 complex formation: This ubiquitination is essential for the formation of the DHX33-NLRP3 complex, a critical step in inflammasome assembly .
Inflammasome activation: The assembled NLRP3 inflammasome activates caspase-1, leading to the processing and secretion of pro-inflammatory cytokines IL-1β and IL-18 .
Requirement across stimuli: TRIM33 is required for inflammasome activation in response to various RNA stimuli, including synthetic dsRNA (poly I:C), bacterial RNA, and viral RNA (reovirus) .
Human relevance: Knockdown experiments in both THP-1-derived macrophages and human monocyte-derived macrophages demonstrate that TRIM33 is essential for dsRNA-induced NLRP3 inflammasome activation in human cells .
This mechanism positions TRIM33 as a key regulator of innate immune responses to nucleic acids derived from pathogens, with potential implications for understanding and treating inflammatory disorders.
TRIM33 serves as a critical regulator of dendritic cell (DC) differentiation and maintenance through dual molecular mechanisms:
Essential for DC differentiation: TRIM33 deletion in Trim33fl/fl Cre-ERT2 mice significantly impairs DC differentiation from hematopoietic progenitors at multiple developmental stages .
Promotion of Irf8 transcription: TRIM33 positively regulates the transcription of Irf8, a key transcription factor essential for the fate specification of conventional dendritic cell subset 1 (cDC1s) .
Transcriptional mechanism: TRIM33 maintains adequate levels of CDK9 and Ser2 phosphorylated RNA polymerase II (S2 Pol II) at Irf8 gene sites, promoting its transcriptional activation .
Prevention of apoptosis: Concurrently, TRIM33 prevents apoptosis of DCs and their progenitors by directly suppressing PU.1-mediated transcription of Bcl2l11 (encoding the pro-apoptotic protein Bim) .
Maintenance of DC homeostasis: Through this dual regulation of differentiation and survival signals, TRIM33 maintains DC homeostasis, ensuring adequate numbers of functional DCs in the immune system .
Broad transcriptional effects: TRIM33 deficiency leads to downregulation of multiple genes associated with DC differentiation in hematopoietic progenitors .
This coordinated regulation of both positive (differentiation-promoting) and negative (apoptosis-inhibiting) signals illustrates how TRIM33 orchestrates complex developmental processes in the immune system, with potential implications for DC-based immune interventions and therapies.
Researchers employ several complementary methods to study TRIM33's interactions with DNA:
Chromatin Immunoprecipitation followed by sequencing (ChIP-seq):
Primary approach for genome-wide mapping of TRIM33 binding sites
Typically uses validated antibodies such as Bethyl Laboratories A301-060A
Requires appropriate controls (Input DNA) for accurate peak calling
Analysis using programs like MACS to identify significant enrichments
Has revealed TRIM33's preference for binding near transcription start sites
ChIP followed by quantitative PCR (ChIP-qPCR):
Analysis of binding to repetitive elements:
Enhancer deletion experiments:
DNA affinity precipitation assays:
Uses biotinylated DNA fragments containing TRIM33 binding sites
Can identify co-binding factors and assess binding specificity
These approaches collectively provide a comprehensive understanding of TRIM33's genomic binding patterns and their functional significance.
Multiple approaches have been established for manipulating TRIM33 expression in experimental systems:
RNA interference (RNAi) techniques:
Short hairpin RNA (shRNA): Stable knockdown has been achieved in THP-1-derived macrophages using at least two distinct TRIM33-targeting shRNAs with good knockdown efficiency
Small interfering RNA (siRNA): Effective in human primary monocyte-derived macrophages (hPMDM), with knockdown efficiency typically exceeding 60% reduction in mRNA levels
Advantage: Relatively simple to implement, can achieve partial knockdown for dose-dependent studies
CRISPR-Cas9 genome editing:
Complete knockout: Generation of TRIM33-null cell lines or animal models
Conditional knockout: Systems like floxed alleles combined with inducible Cre recombinase (e.g., Trim33fl/fl Cre-ERT2 mice)
Knock-in approaches: Creating tagged versions of TRIM33 at endogenous loci
Advantage: Complete elimination of protein, avoiding off-target effects of RNAi
Overexpression systems:
Transfection of expression vectors coding for HA- and Myc-tagged TRIM33 proteins
Generation of truncation constructs to study domain-specific functions
Site-directed mutagenesis to create specific mutants (e.g., RING domain mutants to disrupt E3 ligase activity)
Advantage: Allows structure-function analysis and rescue experiments
Pharmacological approaches:
AlphaScreen assays have been developed for TRIM33 PHD-BRD cassettes, enabling high-throughput screening to identify compounds that modulate TRIM33 function
Novel ligands for TRIM33 have been identified through screening approaches
Advantage: Potential for temporal control and therapeutic development
Each approach has specific advantages depending on the research question, with combinations often providing the most robust evidence for TRIM33's functions.
Researchers employ multiple complementary techniques to investigate TRIM33's E3 ubiquitin ligase activity:
In vitro ubiquitination assays:
Reconstitution of ubiquitination reaction with purified components (E1, E2, TRIM33, substrate, ubiquitin, ATP)
Western blotting detection of ubiquitinated substrate products
Allows detailed biochemical characterization of enzyme kinetics and specificity
Cell-based ubiquitination assays:
Site-directed mutagenesis approaches:
Mass spectrometry-based proteomics:
Identification of ubiquitination sites and linkage types
Quantitative analysis of ubiquitination levels
Global profiling of ubiquitination changes upon TRIM33 manipulation
Functional readouts:
Assessing downstream consequences of TRIM33-mediated ubiquitination
For DHX33 ubiquitination: measuring inflammasome activation via IL-1β and IL-18 production
Testing the effects of ubiquitination site mutations on functional outcomes
Demonstrates that TRIM33's E3 ligase activity is essential for NLRP3 inflammasome activation in response to cytosolic dsRNA
These techniques collectively enable comprehensive characterization of TRIM33's E3 ligase activity, substrate specificity, and the functional consequences of this post-translational modification.
When designing experiments to study TRIM33 in primary cells, researchers should consider these critical factors:
Cell type selection and relevance:
TRIM33 functions are context-dependent, with distinct roles in different cell types
B lymphoblasts: Focus on anti-apoptotic functions and enhancer regulation
Macrophages: Consider inflammasome activation and innate immune responses
Dendritic cells: Examine differentiation pathways and Irf8 regulation
Choose cell types relevant to the specific TRIM33 function being investigated
Isolation and culture conditions:
Genetic manipulation approaches:
Functional readouts:
Stimulation conditions:
Controls and validation:
Careful consideration of these factors will enhance the reliability and physiological relevance of TRIM33 studies in primary cells.
TRIM33 uniquely combines transcriptional regulatory functions with E3 ubiquitin ligase activity, raising complex questions about how these activities are coordinated in specific cellular contexts:
In the context of innate immunity, TRIM33's E3 ligase function is prominent, ubiquitinating the RNA sensor DHX33 to facilitate inflammasome activation . This post-translational modification is critical for the formation of the DHX33-NLRP3 complex. Simultaneously, TRIM33 likely regulates the transcription of genes involved in immune responses, potentially creating a coordinated program of both immediate (ubiquitination-mediated) and sustained (transcription-dependent) responses to pathogens.
Methodological approaches to dissect these integrated functions include:
Domain-specific mutations that selectively disrupt either transcriptional cofactor function (PHD/BRD domains) or E3 ligase activity (RING domain)
Temporal analysis using synchronized cell systems and time-course measurements of both ubiquitination events and transcriptional changes
Proteomics and transcriptomics integration to correlate ubiquitination targets with transcriptional profiles
Proximity labeling approaches to identify context-specific TRIM33 interactors in different cellular compartments
Understanding this functional integration could provide insights into how TRIM33 coordinates complex cellular processes and how its dysregulation contributes to disease states.
TRIM33 has emerged as a regulator of specific classes of repetitive elements in the genome, raising important questions about its role in genome stability and regulation:
ChIP-seq analysis has revealed that TRIM33 shows remarkable enrichment (nearly seven-fold) at RLTR10B elements and to a lesser extent (two-fold) at RLTR10B2 elements . These are specific classes of long terminal repeat (LTR) retrotransposons in the rodent genome. This selective binding suggests TRIM33 may function to silence these potentially mobile genetic elements.
The mechanism of this regulation likely involves TRIM33's ability to function as a transcriptional repressor, potentially recruiting histone-modifying enzymes or other silencing machinery to these repetitive elements. This regulation may be particularly important in germ cells or during early development, when transposon silencing is critical for genomic integrity.
Key research questions in this area include:
Is TRIM33's binding to repetitive elements conserved across species, particularly in humans?
What is the consequence of TRIM33 loss on the expression and mobilization of these elements?
Does TRIM33 cooperate with other known regulators of repetitive elements such as KRAB-ZFPs or TRIM28?
How is TRIM33 specifically recruited to certain classes of repetitive elements but not others?
Does the E3 ligase activity of TRIM33 contribute to its function in repetitive element regulation?
Methodological approaches to address these questions include:
RNA-seq and small RNA sequencing to assess repetitive element expression
ChIP-seq for histone modifications at TRIM33-bound repetitive elements
Long-read sequencing to detect potential transposition events
CRISPR screens to identify cofactors in TRIM33-mediated repetitive element regulation
Understanding TRIM33's role in repetitive element regulation could provide insights into fundamental mechanisms of genome defense and potential connections to diseases associated with dysregulated transposable elements.
Given TRIM33's roles in multiple cellular processes, several therapeutic approaches could be developed to target it in disease contexts:
Small molecule inhibitors of the PHD-BRD cassette:
AlphaScreen assays have been developed for the tandem PHD-BRD cassettes of TRIM33α and TRIM33β
High-throughput screening of approximately 1700 compounds has identified novel ligands for TRIM33
These compounds could disrupt TRIM33's interaction with modified histones, potentially modulating its transcriptional regulatory functions
Such inhibitors could be therapeutically relevant in B cell leukemia, where TRIM33 prevents apoptosis
E3 ligase activity modulators:
Compounds targeting TRIM33's RING domain could modulate its E3 ligase activity
Inhibition could be beneficial in inflammatory conditions by reducing NLRP3 inflammasome activation
Alternatively, PROTAC (Proteolysis-Targeting Chimera) approaches could exploit TRIM33's E3 ligase activity to target disease-relevant proteins for degradation
Protein-protein interaction disruptors:
Gene therapy approaches:
In contexts where TRIM33 is dysregulated, gene therapy to restore normal expression levels
CRISPR-based approaches to correct mutations in TRIM33 or its regulatory elements
Immunotherapeutic strategies:
Development of these therapeutic approaches requires careful consideration of TRIM33's context-dependent functions to achieve specificity and minimize off-target effects. The identification of novel ligands for the TRIM33 PHD-BRD cassette represents a promising first step toward TRIM33-targeted therapeutics .
TRIM33 functions at the intersection of epigenetic regulation and transcriptional control, with its activity profoundly influenced by the epigenetic landscape:
TRIM33 recognizes specific histone modifications through its specialized domains - the PHD domain binds methylated lysine residues (particularly H3K9Me3), while the bromodomain recognizes acetylated lysine residues (notably H3K18Ac) . This recognition is enhanced when both modifications are present, as demonstrated by the preferential binding to dual-modified H3 1-27K9Me3K18Ac peptides .
This epigenetic reading capability allows TRIM33 to:
Respond to cell type-specific epigenetic patterns:
Integrate developmental signals:
Respond to cellular stress or environmental stimuli:
Stimuli like cytosolic RNA can trigger signaling cascades that alter the epigenetic landscape
TRIM33 may relocalize to different genomic regions following such epigenetic remodeling
This mechanism could link environmental sensing to transcriptional reprogramming
Contribute to disease-specific epigenetic states:
Methodological approaches to investigate these relationships include:
Integrated ChIP-seq for TRIM33 and various histone modifications
Single-cell approaches to capture heterogeneity in epigenetic states
CRISPR-based epigenetic editing to manipulate specific modifications and assess TRIM33 binding
Development of histone mimetics to probe TRIM33's epigenetic reading specificity
Understanding how epigenetic contexts influence TRIM33 function could provide insights into both normal development and disease pathogenesis.
Beyond its established functions in transcriptional regulation and ubiquitination, TRIM33 appears to participate in several non-canonical cellular processes that merit further investigation:
Regulation of non-coding RNAs:
The TRIM33 gene sequence shows alignment with 97 transcripts of long non-coding RNA genes and other non-coding elements
This suggests potential regulatory relationships between TRIM33 and the non-coding transcriptome
TRIM33 might modulate non-coding RNA expression or, conversely, be regulated by non-coding RNAs
This represents an emerging area for investigation using RNA immunoprecipitation and functional studies
Control of repetitive element expression:
TRIM33 shows remarkable enrichment at specific classes of endogenous retroelements, particularly RLTR10B elements
This suggests a role in genome defense against potentially mobile genetic elements
The functional consequences of this binding and potential implications for genome stability require further exploration
Intersection with RNA sensing pathways:
TRIM33's role in cytosolic RNA-induced inflammasome activation connects it to cellular RNA sensing mechanisms
This raises questions about whether TRIM33 might participate in other RNA-related processes, such as RNA processing, export, or quality control
The ubiquitination of RNA-binding proteins like DHX33 by TRIM33 could have broader implications for RNA metabolism
Potential roles in metabolism:
E3 ligases often regulate metabolic enzymes and pathways through ubiquitination
TRIM33's cell type-specific functions might extend to metabolic regulation, particularly in immune cells where metabolic reprogramming is critical for function
Involvement in cellular stress responses:
Investigating these non-canonical functions requires integrative approaches combining proteomics, transcriptomics, and functional genomics, potentially revealing unexpected roles for TRIM33 in cellular homeostasis and disease.
The TRIM33 gene is located on chromosome 1p13.2 in humans . The protein structure includes:
Three alternatively spliced transcript variants of this gene have been described, although the full-length nature of one variant has not been determined .
TRIM33 functions primarily as a transcriptional corepressor and an E3 ubiquitin-protein ligase . It plays a crucial role in various biological processes, including:
TRIM33 is expressed in various tissues, including the secondary oocyte, epithelium of the choroid plexus, Brodmann area 23, corpus epididymis, nipple, cerebellar vermis, tibia, and region I of the hippocampus proper . It is also expressed in the genital tubercle, primitive streak, tail of the embryo, ciliary body, zygote, trigeminal ganglion, iris, and retinal pigment epithelium .
TRIM33 has been implicated in various diseases, particularly in cancer. It functions as a tumor suppressor and is involved in the regulation of hematopoietic stem cell aging . Its role in protein ubiquitination and transcriptional regulation makes it a potential target for therapeutic interventions.