Recombinant Mouse Serine/threonine-protein phosphatase 6 regulatory subunit 1 (Ppp6r1), partial, refers to a genetically engineered fragment of the Ppp6r1 protein derived from a mouse. Ppp6r1, or protein phosphatase 6 regulatory subunit 1, is a regulatory component of protein phosphatase 6 (PP6) . PP6 is a serine/threonine phosphatase involved in various cellular processes .
Ppp6r1 likely functions as a scaffolding subunit within the PP6 complex . It may participate in the PP6-mediated dephosphorylation of NFKBIE, which opposes its degradation in response to TNF-alpha .
Ppp6r1 is predicted to enable protein phosphatase regulator activity and small GTPase binding activity . As a regulatory subunit, Ppp6r1 helps target protein phosphatase-6 to specific substrates and locations within the cell .
Various substances can affect the expression and methylation of Ppp6r1 . Some examples include:
2-palmitoylglycerol increases expression.
3,3',5,5'-tetrabromobisphenol A and decabromodiphenyl ether decrease expression.
3,4-methylenedioxymethamphetamine, cobalt dichloride, copper(II) chloride, cyclosporin A, dicrotophos, fenthion, finasteride, flutamide, thiram, titanium dioxide, and urethane increase expression.
4,4'-sulfonyldiphenol and bisphenol A affect methylation.
6-propyl-2-thiouracil, bisphenol A, and valproic acid affect expression.
Acrolein, CGP 52608, ethanol and maneb have multiple interactions.
Beta-lapachone and lead(0) decrease expression.
Chlordecone and folic acid decrease expression.
FR900359 increases phosphorylation.
Temozolomide decreases expression.
Titanium dioxide decreases methylation.
Regulatory subunit of protein phosphatase 6 (PP6). It may function as a scaffolding PP6 subunit and is involved in PP6-mediated dephosphorylation of NFKBIE, counteracting its degradation in response to TNF-alpha.
Ppp6r1 (also known as SAPS1) is a regulatory subunit for the protein phosphatase-6 catalytic subunit (PP6c). Within the PP6 complex, it modulates enzymatic activity through three primary mechanisms: restricting substrate specificity, recruiting substrates to the complex, and determining the intracellular localization of the holoenzyme . The SAPS domain within Ppp6r1 mediates interaction with the catalytic subunit, while other regions facilitate binding to specific substrates like DNA-PK .
Mouse Ppp6r1 shares significant sequence homology with human PPP6R1, with specific regions showing particularly high conservation. For example, the amino acid regions 364-456 in humans show approximately 84% identity with mouse orthologs, while regions 782-864 show approximately 70% identity .
Unlike some phosphatase complexes that require only catalytic and regulatory subunits, the PP6 holoenzyme follows a more complex assembly pattern involving multiple interacting components. The PP6 complex consists of:
A catalytic subunit (PP6c)
One of three SAPS domain-containing regulatory subunits (Ppp6r1/SAPS1, Ppp6r2/SAPS2, or Ppp6r3/SAPS3)
One of three ankyrin repeat domain proteins (ANKRD28, ANKRD44, or ANKRD52)
This tripartite structure allows for highly specific targeting and regulation. PP6c interacts directly with Ppp6r1 through its active site, and mutations in PP6c's catalytic domain (such as H114A) can completely disrupt this association, suggesting that subtle conformational changes in PP6c are detected by the SAPS domain of Ppp6r1 .
When validating antibody specificity for mouse Ppp6r1, a multi-step approach is recommended:
Blocking experiments: Use recombinant protein control fragments corresponding to the antibody recognition region. Pre-incubate the antibody with a 100x molar excess of the protein fragment control for 30 minutes at room temperature before proceeding with immunohistochemistry (IHC), immunocytochemistry (ICC), or Western blotting (WB) .
siRNA knockdown validation: Implement siRNA-mediated knockdown of Ppp6r1 (targeting >80% reduction) alongside non-targeting siRNA controls. Compare antibody reactivity in both samples via Western blotting and immunoprecipitation to confirm specificity .
Cross-reactivity assessment: Evaluate potential cross-reactivity with related proteins, particularly other SAPS family members (SAPS2/Ppp6r2 and SAPS3/Ppp6r3), which share structural similarities with Ppp6r1.
Orthogonal methods: Combine antibody-based detection with mass spectrometry validation or recombinant expression systems to verify target identity.
To investigate how Ppp6r1 recruits substrates to the PP6 complex:
Co-immunoprecipitation studies: Implement bidirectional co-IP experiments using tagged versions of Ppp6r1 and potential substrates. This approach has successfully identified DNA-PK as a Ppp6r1 substrate through direct interaction .
Domain mapping experiments: Create a series of truncation mutants to identify specific binding domains within Ppp6r1. Research has revealed that Ppp6r1 contains two distinct regions that mediate interaction with substrates: one dominant binding site and a secondary supporting site .
Phosphoproteomic analysis: Following Ppp6r1 depletion or overexpression, implement quantitative phosphoproteomics to identify differentially phosphorylated proteins, which may represent direct or indirect substrates.
Proximity-based labeling: Use BioID or APEX2 fusion proteins to identify proteins in close proximity to Ppp6r1 within cells, potentially revealing previously unknown interaction partners.
Ppp6r1 plays a critical role in DNA damage response through its interaction with DNA-dependent protein kinase (DNA-PK):
Recruitment mechanism: Ppp6r1 acts as a bidentate anchor that bridges PP6c to DNA-PK independently of PP6c's phosphatase activity. This interaction involves two distinct binding regions within Ppp6r1, with one site serving as the dominant interface and the other providing supporting contact .
Functional significance: The PP6 complex is required for proper DNA-PK activation following ionizing radiation. Knockdown experiments demonstrate that reduced Ppp6r1 levels significantly diminish the association between PP6c and DNA-PK, impairing the cellular response to DNA damage .
Therapeutic implications: The DNA-PK-Ppp6r1 interface represents a potential drug target for radiosensitizing cells, as disruption of this interaction could inhibit DNA-PK activation and enhance sensitivity to radiotherapy .
Recent research has uncovered Ppp6r1's involvement in cytokine signaling pathways:
TNFα signaling modulation: The PP6 complex, which includes Ppp6r1, has been identified as a component of the TNF receptor signaling complex I. Loss of PP6 activity protects cells from TNFα-mediated cell death .
Mechanistic action: PP6 negatively modulates LUBAC-mediated M1-ubiquitination of key signaling molecules like RIPK1 and c-FLIP. This modulation affects cell fate decisions in response to TNFα stimulation .
Pathological relevance: Inactivating mutations in PP6 components (including regulatory subunits) have been associated with cancer progression, particularly in melanoma. These mutations appear to promote cancer cell survival by rendering cells less sensitive to TNFα-induced cell death .
Ppp6r1 has emerged as a regulator of fetal hemoglobin (HbF) production with potential therapeutic implications:
Expression pattern: Unlike PP6c, which is broadly expressed across hematopoietic lineages, Ppp6r1 shows enrichment in erythroid precursors, suggesting lineage-specific functions .
HbF regulation: CRISPR-Cas9-mediated depletion of Ppp6r1 in primary human erythroid cells increases HBG (fetal β-globin) mRNA levels 2-3 fold and nearly doubles the number of HbF-expressing cells .
Mechanistic pathway: The PP6 complex appears to regulate HbF production partly through modulating levels of BCL11A, a known HbF repressor. Depletion of PP6 components reduces BCL11A levels by approximately 50%, while leaving other HbF regulators unchanged .
Therapeutic potential: In sickle cell disease patient-derived cells, PP6 component depletion leads to robust HbF induction and reduces cell sickling in vitro by up to 60%. Xenotransplantation studies have demonstrated sustained HbF induction (~4-fold) at 16 weeks post-transplant .
Several experimental systems have proven effective for investigating Ppp6r1's role in erythroid development:
HUDEP2 cell line: This system has been successfully used in domain-focused CRISPR-Cas9 screens targeting serine/threonine phosphatases, leading to the identification of PP6 components as HbF regulators .
Primary human erythroid cultures: Ex vivo differentiation of CD34+ hematopoietic stem/progenitor cells provides a physiologically relevant system for studying Ppp6r1 function during erythropoiesis. This model has confirmed dose-dependent effects of PP6 component depletion on HbF levels .
Patient-derived sickle cell disease models: Primary cells from sickle cell disease patients offer the ability to assess both molecular changes (HbF induction) and functional outcomes (reduced sickling) .
NBSGW xenotransplantation model: This immunodeficient mouse model supports human erythropoiesis and has been used to demonstrate sustained in vivo effects of PP6 component depletion on HbF expression over extended periods (16+ weeks) .
Recombinant Ppp6r1 fragments serve as valuable tools for detailed interaction studies:
Competition assays: Defined fragments can be used to compete with full-length protein for binding partners, helping to map specific interaction domains. For example, recombinant fragments corresponding to amino acids 364-456 or 782-864 can help determine which regions mediate binding to different partners .
Pull-down experiments: Immobilized recombinant fragments can capture interaction partners from cell lysates, allowing identification of proteins that bind to specific domains of Ppp6r1.
Structural studies: Well-defined recombinant fragments enable crystallography or cryo-EM studies to determine atomic-level details of protein interactions.
Antibody validation: Control fragments are essential for confirming antibody specificity through blocking experiments, where pre-incubation with the fragment prevents antibody binding in subsequent applications .
Identifying novel substrates regulated by Ppp6r1-containing phosphatase complexes requires sophisticated methodological approaches:
Phosphoproteomic analysis with temporal resolution: Implement quantitative phosphoproteomics following acute depletion of Ppp6r1 (e.g., using dTAG-based systems) to identify rapidly changing phosphosites, which are more likely to represent direct substrates .
Substrate-trapping mutants: Generate phosphatase-dead mutants of PP6c that can still bind substrates but not dephosphorylate them, creating stable enzyme-substrate complexes that can be isolated and identified.
Proximity-dependent labeling combined with phosphoproteomics: Fuse BioID or APEX2 to Ppp6r1 to label proteins in its vicinity, then analyze the phosphorylation status of these proteins to identify potential substrates.
In vitro dephosphorylation assays: Use purified PP6 holoenzyme containing Ppp6r1 to dephosphorylate candidate substrates in vitro, confirming direct enzymatic action.
Bioinformatic prediction: Apply machine learning algorithms trained on known phosphatase substrates to predict new candidates, which can then be validated experimentally.
When working with recombinant mouse Ppp6r1, researchers should assess:
Purity assessment: Use SDS-PAGE with Coomassie staining to verify >90% purity and confirm the expected molecular weight.
Functional validation: Implement binding assays with known interaction partners (e.g., PP6c) to confirm that the recombinant protein maintains its biological activity.
Structural integrity: Use circular dichroism spectroscopy to evaluate secondary structure content and proper folding.
Aggregation analysis: Employ dynamic light scattering and size-exclusion chromatography to detect potential protein aggregation.
Endotoxin testing: For preparations intended for cell culture or in vivo studies, verify endotoxin levels are below 1 EU/mg protein.
When facing reproducibility challenges in Ppp6r1 research:
Cell cycle variability: Phosphatase substrate interactions often vary throughout the cell cycle. Synchronize cells or sort them by cell cycle phase to reduce variability, as PP6 has established roles in cell cycle regulation .
Post-translational modification status: Check for potential phosphorylation or other modifications on Ppp6r1 itself, which might regulate its interaction capacity.
Holoenzyme composition: Verify the expression levels of other PP6 complex components, as variations in their abundance can affect substrate recruitment and specificity.
Buffer conditions: Optimize immunoprecipitation buffer conditions, as ionic strength and detergent composition can significantly impact the stability of protein-protein interactions.
Technical approach diversity: Implement multiple complementary techniques (co-IP, proximity labeling, in vitro binding assays) to build a stronger case for specific interactions.
Recent findings suggest promising therapeutic applications for Ppp6r1 in hemoglobinopathies:
Sickle cell disease therapy: Targeted inhibition of the PP6 complex, potentially through Ppp6r1-specific approaches, could provide a novel strategy for increasing fetal hemoglobin levels in patients with sickle cell disease. This approach has shown efficacy in reducing sickling in patient-derived cells by up to 60% .
Erythroid-selective targeting: The enriched expression of Ppp6r1 in erythroid precursors offers an opportunity for lineage-specific therapeutic targeting, potentially minimizing off-target effects in other cell types .
Gene therapy approaches: CRISPR-Cas9 targeting of Ppp6r1 in hematopoietic stem cells could provide a long-term solution for hemoglobinopathies, as demonstrated by sustained HbF induction in xenotransplantation models .
Combination therapies: Modulating Ppp6r1 activity could potentially synergize with other HbF-inducing agents, providing more robust therapeutic outcomes than single-agent approaches.
The bidentate nature of Ppp6r1's interaction with substrates offers unique therapeutic opportunities:
Small molecule inhibitors: The presence of two distinct binding regions on Ppp6r1 that mediate interaction with substrates like DNA-PK provides multiple potential targets for small molecule inhibitor development .
Peptide-based disruptors: Synthetic peptides mimicking either the dominant or supporting binding sites could compete with natural interactions and selectively disrupt Ppp6r1-substrate binding.
Allosteric modulators: Compounds that bind to Ppp6r1 and induce conformational changes could alter the presentation of one or both binding interfaces, modulating rather than completely blocking interactions.
Specificity engineering: The dual binding mechanism potentially allows for development of more specific inhibitors that target unique combinations of the dominant and supporting binding sites, reducing off-target effects.