Recombinant Mouse Protein polybromo-1 (Pbrm1), partial, refers to a genetically engineered version of the mouse polybromo-1 protein, which is a crucial component of the PBAF (Polybromo, Brg1-Associated Factors) chromatin remodeling complex. This complex plays a pivotal role in regulating gene expression by altering chromatin structure, thereby controlling DNA accessibility and transcriptional activity . The partial recombinant protein typically includes specific domains or regions of the full-length protein, which are essential for its function in chromatin targeting and remodeling.
Polybromo-1 (Pbrm1) is characterized by its unique structure, which includes six tandem bromodomains, two bromo-adjacent homology (BAH) domains, and a high-mobility group (HMG) domain . The bromodomains are critical for binding to acetylated histones, which helps in targeting the PBAF complex to specific chromatin sites . The BAH domains facilitate protein-protein interactions, while the HMG domain interacts with nucleosomal DNA .
| Domain | Function |
|---|---|
| Bromodomains | Bind acetylated histones, targeting chromatin sites |
| BAH Domains | Protein-protein interactions |
| HMG Domain | Interacts with nucleosomal DNA |
Pbrm1 is frequently mutated in clear cell renal cell carcinoma (ccRCC), where it acts as a tumor suppressor . Mutations in Pbrm1 are believed to be early events in carcinogenesis, affecting gene expression related to cellular adhesion, metabolism, and cell division . Additionally, Pbrm1 has been implicated in myeloid leukemia progression, where its loss promotes leukemogenesis by affecting immune regulation and interferon signaling .
Recent studies have highlighted the importance of individual bromodomains in Pbrm1 for chromatin association and tumor suppression. For instance, bromodomains 2 and 4 are crucial for binding to histone H3 acetylated at lysine 14, which is essential for cell proliferation and gene expression . The development of selective inhibitors targeting Pbrm1 bromodomains could provide insights into its role in cancer pathogenesis and immunotherapy .
| Bromodomain | Function | Importance |
|---|---|---|
| BD2 and BD4 | Bind H3K14ac | Critical for cell proliferation and gene expression |
| BD1, BD5, BD6 | Enhance binding | Contribute to chromatin targeting |
| BD3 | Attenuates binding | Modulates chromatin interaction |
Polybromo-1: the chromatin targeting subunit of the PBAF complex - PMC2646799
PBRM1 bromodomains associate with RNA to facilitate chromatin... - NAR
PBRM1 Regulates the Expression of Genes Involved in Metabolism and Cell Adhesion in Renal Clear Cell Carcinoma - PMC4839679
Recombinant PBRM1 (613-734) protein bromodomain - Active Motif
The RBPJ/DAPK3/UBE3A signaling axis induces PBRM1... - Nature
PBRM1 bromodomains variably influence nucleosome interactions and cellular function - PMC6120218
PBRM1 Gene - GeneCards
Pbrm1 polybromo 1 [ (house mouse)] - NCBI
In vivo CRISPR/Cas9 screening identifies Pbrm1 as a regulator of myeloid leukemia development in mice - PMC10506108
Selective and Cell-Active PBRM1 Bromodomain Inhibitors Discovered through NMR Fragment Screening - PMC9630929
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes. We will fulfill requests whenever possible.
Note: All protein shipments include standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during production. To request a specific tag, please inform us, and we will prioritize its development.
STRING: 10090.ENSMUSP00000107727
UniGene: Mm.27913
Pbrm1 (also known as BAF180) contains six tandem bromodomains (BrDs), two bromo-adjacent homology domains (BAH), and a high-mobility group (HMG). This multi-domain architecture enables Pbrm1 to coordinate several essential functions within the PBAF complex . The bromodomains specifically recognize and bind to acetylated lysine residues on histone tails, with different bromodomains targeting distinct acetylation patterns. The BAH domains function as protein-interaction modules, while the HMG domain binds nucleosomal DNA .
This multifunctional structure allows Pbrm1 to target the PBAF complex to specific chromatin sites defined by discrete nucleosome acetylation patterns, recruit additional regulatory proteins, and directly participate in altering histone-DNA interactions to control genetic functions . Quantitative screening has revealed that several Pbrm1 bromodomains target specific acetyllysines within histone tail regions, suggesting that Pbrm1 recognizes a combinatorial histone acetylation code .
Pbrm1 expression can be studied using mouse models such as the Pbrm1 tm1a(EUCOMM)Wtsi ES cells. These models allow for β-galactosidase staining to determine Pbrm1 expression patterns across different tissues. For tissue-specific studies, various Cre-driver mouse lines can be used, including Sglt2-Cre, Villin-Cre, and Pax8-Cre mice, which enable conditional deletion of Pbrm1 in specific cell types .
Researchers have found that Pbrm1 is widely expressed across multiple tissues but has particularly important functions in renal cells. The studies with Pax8-Cre and Ksp-Cre driver lines have been instrumental in understanding Pbrm1's role in kidney development and renal cell carcinoma formation .
Recombinant mouse Pbrm1 protein is valuable for numerous experimental applications:
Pbrm1 deficiency significantly impacts IFNγ signaling pathways and shapes the tumor immune microenvironment. Mechanistically, Pbrm1 loss reduces the binding of brahma-related gene 1 (BRG1) to the IFNγ receptor 2 (Ifngr2) promoter . This molecular alteration leads to decreased STAT1 phosphorylation and subsequent reduction in the expression of IFNγ target genes .
The consequences of this disrupted signaling are profound. Analysis across three independent patient cohorts and murine pre-clinical models has demonstrated that PBRM1 loss is consistently associated with:
A less immunogenic tumor microenvironment (TME)
Upregulated angiogenesis
In mouse models, Pbrm1-deficient Renca subcutaneous tumors showed greater resistance to immune checkpoint blockade compared to Pbrm1-intact tumors . This has significant implications for immunotherapy strategies in cancers with PBRM1 mutations.
Creating effective Pbrm1 knockout mouse models requires careful consideration of several methodological factors:
Selection of appropriate ES cells: Pbrm1 tm1a(EUCOMM)Wtsi ES cells have been successfully used for generating germline transmission .
Breeding strategy: Male chimeras should be mated with C57BL/6NTac female mice to generate germline transmission. Heterozygous mice can then be used for β-galactosidase staining to confirm Pbrm1 expression patterns .
Conditional knockout approach: Floxed Pbrm1 mice can be generated by mating Pbrm1-LacZ/+ mice with FLP mice to excise the LacZ element .
Selection of Cre drivers: Different tissue-specific Cre drivers yield distinct phenotypes:
Co-targeting considerations: When studying renal cell carcinoma, co-targeting Vhl with Pbrm1 produces more robust tumor models than targeting either gene alone .
Pbrm1 mutations have emerged as potential biomarkers for immunotherapy response across multiple cancer types. Comprehensive analysis of clinical data reveals that patients with PBRM1 mutations exhibit:
Improved progression-free survival (PFS) (HR = 0.51, 95% CI: 0.28–0.95, p = 0.030)
Higher objective response rates (ORR) (47.92% vs. 28.21%, p = 0.0044)
Better disease control rates (DCR) (72.92% vs. 47.53%, p = 0.0008)
These benefits have been observed across multiple cancer types. In non-small cell lung cancer specifically, patients with PBRM1 mutations demonstrated:
Significantly longer PFS (HR = 0.268, 95% CI: 0.084–0.854, p = 0.04)
Higher ORR (55.56% vs. 20.00%, p = 0.027)
Gene set enrichment analysis (GSEA) reveals that PBRM1 mutations are closely related to immune efficacy and immune microenvironment, including enhanced killer cell-mediated immunity regulation, cell cytokine production, CD8+ T-cell activation, and MHC protein binding processes .
Evaluating the binding specificity of Pbrm1's bromodomains requires a multifaceted experimental approach:
Effective comparison of phenotypes between different Pbrm1 knockout mouse models requires standardized methodology:
Consistent genetic background: Use mice with identical or closely related genetic backgrounds to minimize variation unrelated to Pbrm1 status.
Standardized phenotypic assessment: Develop comprehensive evaluation protocols for:
Cell-of-origin tracking: Use lineage tracing approaches (e.g., with Rosa26-CAG-loxP-stop-loxP-tdTomato mice) to identify the cellular origins of arising tumors .
Comparison across different Cre-drivers: When comparing models with different Cre-drivers (e.g., Ksp-Cre vs. Pax8-Cre), document:
Molecular and signaling pathway analysis: Assess activation of key signaling pathways (e.g., mTORC1) across different models to identify conserved mechanisms .
To investigate Pbrm1's role in immune checkpoint blockade resistance, researchers should consider these experimental design elements:
In vivo tumor models:
Treatment protocols:
Immune profiling:
Mechanistic investigations:
Clinical correlation:
Pbrm1/PBRM1 mutation status has significant potential as a predictive biomarker for immunotherapy response based on multiple lines of evidence:
To determine the functional consequences of specific Pbrm1 mutations, researchers can employ these experimental approaches:
Domain-specific mutation analysis:
Create a panel of recombinant Pbrm1 proteins with mutations in specific domains (bromodomains, BAH domains, HMG domain)
Test each mutant's ability to bind acetylated histones, interact with other PBAF components, and remodel chromatin
Compare naturally occurring cancer mutations with engineered mutations
Cell-based functional assays:
Chromatin remodeling activity assessment:
In vitro nucleosome remodeling assays with reconstituted PBAF complexes containing wild-type or mutant Pbrm1
Measure changes in chromatin accessibility, nucleosome positioning, and DNA exposure
Protein-protein interaction analysis:
Immunoprecipitation followed by mass spectrometry to identify differential interactors
Proximity labeling techniques (BioID, APEX) to map protein interaction networks in living cells
Yeast two-hybrid screening to identify affected protein interactions
In vivo modeling:
Investigating the cooperation between Pbrm1 and other tumor suppressors in renal cell carcinoma requires systematic approaches:
Combinatorial genetic modeling:
Molecular pathway analysis:
Perform comprehensive multi-omic profiling (genomics, transcriptomics, proteomics, metabolomics)
Identify convergent and divergent pathway alterations
Map chromatin landscape changes using ChIP-seq and ATAC-seq
Analyze effects on specific pathways (e.g., mTORC1, which is differentially activated based on which tumor suppressors are lost)
Cell-of-origin studies:
Therapeutic response assessment:
Clinical correlation: