PPIL2 suppresses breast cancer metastasis by:
Inhibiting epithelial-mesenchymal transition (EMT) through SNAI1 downregulation :
Promoting SNAI1 ubiquitination and proteasomal degradation (half-life reduced from 4.5 hr to 2.1 hr) .
PPIL2 regulates homologous recombination (HR) repair:
Mechanism | Effect on HR | Key Partners |
---|---|---|
CtIP ubiquitination | Inhibits HR efficiency | ZNF830 (recruitment factor) |
PLK1-mediated phosphorylation | Enhances HR repair | BLM helicase |
PPIL2 knockdown increases HR efficiency by 37% while reducing non-homologous end joining (NHEJ) by 22% in U2OS cells .
PPIL2 interacts with multiple partners through distinct domains:
Breast cancer: PPIL2 expression inversely correlates with metastasis-free survival (HR = 0.62, p < 0.01) .
DNA repair targeting: PLK1-mediated PPIL2 phosphorylation at Ser-154 enhances CtIP activity, suggesting druggable interface .
Recombinant PPIL2 protein (His-tagged) available for in vitro studies (MedChemExpress, Cat# HY-P70295) .
Antibodies validated in immunohistochemistry (Human Protein Atlas) .
PPIL2 shows ubiquitous but variable expression:
Tissue | Expression Level | Notable Localization |
---|---|---|
Brain | High | Hippocampus, cerebral cortex |
Reproductive organs | Moderate | Testis, ovary |
Immune system | Low | Spleen, lymph nodes |
RNA-seq data indicates highest expression in the cerebral cortex (FPKM = 12.3) and lowest in skeletal muscle (FPKM = 2.1) .
MGSSHHHHHH SSGLVPRGSH MGKRQHQKDK MYITCAEYTH FYGGKKPDLP QTNFRRLPFD HCSLSLQPFV YPVCTPDGIV FDLLNIVPWL KKYGTNPSNG EKLDGRSLIK LNFSKNSEGK YHCPVLFTVF TNNTHIVAVR TTGNVYAYEA VEQLNIKAKN FRDLLTDEPF SRQDIITLQD PTNLDKFNVS NFYHVKNNMK IIDPDEEKAK QDPSYYLKNT NAETRETLQE YKEFKGDEI LAATMKAPEK KKVDKLNAAH YSTGKVSASF TSTAMVPETT EAAAIDEDV LRYQFVKKKG YVRLHTNKGD LNLELHCDLT PKTCENFIRL CKKHYYDGTI FHRSIRNFVI QGGDPTGTGT GGESYWGKPF KDEFRPNLSH TGRGILSMAN SGPNSNRSQF FITFRSCAYL DKKHTIFGRV VGGFDVLTAM ENVESDPKTD RPKEEIRIDA TTVFVDPYEE ADAQIAQERK TQLKVAPETK VKSSQPQAGS QGPQTFRQGV GKYINPAATE QQRKSPQPVP LSPCPRRSPV GVLGTSAPGS SRLPDDH.
PPIL2 (Peptidylprolyl Isomerase Like 2) functions as a U-box ubiquitin ligase that plays multiple roles in cellular processes. Primary research indicates PPIL2 is involved in:
Regulating protein stability through ubiquitination of target proteins, particularly TP53 (p53)
Participating in DNA damage repair pathways, specifically homologous recombination (HR)
Contributing to erythropoiesis (red blood cell formation)
Acting as a downstream effector in the JAK2/STAT5 signaling pathway
To study PPIL2's primary function in your experimental system, consider using knockdown approaches (CRISPR-PPIL2 sgRNA) in relevant cell types (such as CD34+ hematopoietic cells for erythropoiesis studies), followed by assessment of proliferation, differentiation, and specific pathway activities . For DNA repair studies, reporter assays measuring HR efficiency provide a quantitative readout of PPIL2 function .
PPIL2 expression is regulated through multiple mechanisms:
Transcriptional regulation: Evidence shows PPIL2 is a direct transcriptional target of the JAK2/STAT5 pathway. Dual-luciferase reporter assays have confirmed that STAT5 can bind directly to the PPIL2 gene promoter .
Cell cycle-dependent expression: PPIL2 levels fluctuate during the cell cycle, with decreased expression observed during M phase in U2OS cells .
Disease-state regulation: PPIL2 is upregulated in myeloproliferative neoplasms (MPNs), particularly those positive for the JAK2V617F mutation .
To study PPIL2 regulation, methodologically sound approaches include:
Chromatin immunoprecipitation (ChIP) to confirm transcription factor binding to the PPIL2 promoter
Synchronization of cell populations to track expression through cell cycle phases
Comparative expression analysis between normal and disease tissues using qPCR or Western blotting
For reliable detection of PPIL2 in human samples, researchers should consider the following methodological approaches:
Western blotting: Use validated antibodies against PPIL2 with appropriate positive and negative controls. When comparing expression levels across samples, normalization to loading controls (β-actin, GAPDH) is essential.
Immunoprecipitation (IP): Particularly useful for studying PPIL2 interactions with binding partners. IP-mass spectrometry (IP-MS) has successfully identified PPIL2 interacting proteins in erythroid cells .
Immunofluorescence: For subcellular localization studies, PPIL2 can be detected using fluorescence microscopy with appropriate antibodies.
Flow cytometry: For quantitative analysis in hematopoietic cells, intracellular staining protocols can be optimized for PPIL2 detection.
For all detection methods, validation through multiple approaches is recommended, as antibody specificity can be variable. Knockdown controls should be included to confirm signal specificity.
PPIL2 has been shown to regulate TP53 stability through the ubiquitin-proteasome system. Research findings demonstrate:
PPIL2 physically interacts with TP53 in erythroid cells, as confirmed by immunoprecipitation and mass spectrometry (IP-MS) .
PPIL2 catalyzes polyubiquitination of TP53, targeting it for proteasomal degradation. This mechanism was confirmed through ubiquitination assays and treatment with the proteasome inhibitor MG132, which reverses TP53 downregulation by PPIL2 .
This regulatory relationship has significant implications for cancer biology, as TP53 is a critical tumor suppressor.
For researchers investigating this pathway, the following methodological approaches are recommended:
Co-immunoprecipitation experiments with both endogenous and tagged proteins to confirm interaction
In vitro and in vivo ubiquitination assays using purified components
Cycloheximide chase experiments to assess TP53 half-life in PPIL2-manipulated cells
Analysis of downstream TP53 targets using qPCR or proteomic approaches
When designing experiments to study this relationship, careful attention should be paid to cell type selection, as PPIL2-TP53 interactions may vary in different cellular contexts.
PPIL2 has been identified as a regulator of DNA damage repair, specifically affecting homologous recombination (HR). Key research findings indicate:
PPIL2 is recruited to DNA double-strand break (DSB) sites in an ATM-dependent manner, as demonstrated by chromatin immunoprecipitation (ChIP) analysis .
PPIL2 negatively regulates HR repair efficiency. Knockdown of PPIL2 promotes HR, as measured using I-SceI-mediated reporter systems .
PPIL2's effect on HR is CtIP-dependent. When CtIP is depleted, PPIL2 knockdown no longer affects HR repair efficiency .
PPIL2 can ubiquitinate CtIP, a key factor in DNA end resection during HR, potentially affecting its activity or stability.
For researchers investigating PPIL2's role in DNA repair, the following methodological approaches are valuable:
Reporter assays measuring HR, NHEJ, and MMEJ repair pathway activity
Laser micro-irradiation combined with live-cell imaging to track recruitment kinetics
ChIP-seq to map PPIL2 binding at endogenous damage sites
Co-immunoprecipitation and ubiquitination assays to characterize PPIL2-CtIP interaction
When designing experiments, consider cell cycle synchronization approaches, as HR is primarily active during S/G2 phases.
PLK1 (Polo-like kinase 1) has been identified as a key regulator of PPIL2 through phosphorylation. Research findings demonstrate:
PPIL2 physically interacts with PLK1, primarily through PLK1's polo-box domain (PBD) .
PLK1 phosphorylates PPIL2 at multiple sites (14 sites have been identified) .
This phosphorylation appears to reduce PPIL2's ability to ubiquitinate CtIP, thereby promoting HR repair activity.
The interaction between PLK1 and PPIL2 increases in the presence of DNA double-strand breaks .
For researchers investigating this regulatory relationship, recommended methodological approaches include:
In vitro kinase assays to confirm direct phosphorylation
Phospho-specific antibodies or mass spectrometry to track phosphorylation status
Mutational analysis of phosphorylation sites to identify functional significance
Cellular assays measuring HR efficiency with phospho-mimetic or phospho-dead PPIL2 mutants
When designing experiments to study PLK1-PPIL2 interactions, consider the cell cycle context, as PLK1 activity peaks during G2/M phase but also functions during S phase in DNA repair contexts.
For investigating PPIL2's function in erythropoiesis, several experimental models have proven effective:
Human CD34+ hematopoietic stem and progenitor cell (HSPC) culture systems:
Mouse models:
Cell line models:
Erythroleukemia cell lines (K562, HEL) can be used for mechanistic studies
Should be validated with primary cell experiments due to potential aberrant signaling
When using these models, the following methodological considerations are critical:
Assessment of erythroid differentiation via flow cytometry (CD71, CD235a/Glycophorin A)
Cell proliferation and apoptosis measurements
Gene expression profiling at different differentiation stages
Biochemical analysis of JAK2/STAT5 pathway activation
PPIL2 has been identified as a potential contributor to myeloproliferative neoplasm (MPN) pathogenesis, particularly in JAK2V617F-positive disease. Research findings indicate:
PPIL2 is a downstream target of the JAK2/STAT5 pathway, which is constitutively activated in JAK2V617F-positive MPNs .
PPIL2 expression is upregulated in MPN patient samples and in JAK2V617F-positive mouse models .
Loss of Ppil2 ameliorates JAK2V617F-induced myeloproliferative phenotypes including erythrocytosis and splenomegaly, as demonstrated in mouse transplantation experiments .
PPIL2 inhibition using cyclosporin A (CsA) similarly reduces MPN disease manifestations .
For researchers investigating PPIL2's role in MPNs, the following methodological approaches are recommended:
Comparative expression analysis between normal and MPN patient samples
JAK2V617F mouse models with Ppil2 manipulation (knockout, knockdown)
Pharmacological inhibition studies using compounds that target PPIL2
Mechanistic studies examining PPIL2's regulation of TP53 in the context of JAK2V617F signaling
When designing experiments, consider the complex interplay between JAK2/STAT5 signaling, PPIL2 expression, TP53 regulation, and erythroid proliferation/differentiation.
When designing experiments to manipulate PPIL2 expression, researchers should consider multiple approaches based on their experimental goals:
CRISPR-Cas9 mediated knockout:
shRNA/siRNA knockdown:
Inducible systems:
Tet-on/off systems allow temporal control of PPIL2 expression
Particularly valuable for studying dynamic processes
Can help distinguish between developmental and acute effects
Domain-specific mutations:
For studying specific functions (e.g., ubiquitin ligase activity)
Requires knowledge of functional domains
Can be introduced using CRISPR-based knock-in approaches
The choice between these approaches should be guided by:
Cell type (primary cells vs. cell lines)
Temporal requirements (acute vs. chronic depletion)
Experimental readout (biochemical vs. phenotypic)
Need for complete vs. partial loss of function
Given PPIL2's role as a U-box ubiquitin ligase, ubiquitination assays are critical for understanding its function. Optimal experimental design should consider:
In vitro ubiquitination assays:
Require purified components (E1, E2 enzymes, PPIL2, substrate, ubiquitin)
Allow direct assessment of PPIL2's catalytic activity
Can identify specific ubiquitin chain types (K48, K63, etc.)
Western blot detection with substrate-specific antibodies
Cell-based ubiquitination assays:
Controls and variables to consider:
PPIL2 catalytic mutants as negative controls
Varying substrate concentrations to determine enzyme kinetics
Time-course experiments to capture ubiquitination dynamics
Deubiquitinating enzyme inhibitors to prevent reversal of ubiquitination
Specific analysis for TP53 ubiquitination:
IP of TP53 followed by ubiquitin detection
Cycloheximide chase experiments to assess TP53
Analysis of TP53 target gene expression as functional readout
Comparison of wild-type vs. mutant TP53 as substrates
When interpreting results, distinguish between mono-ubiquitination and poly-ubiquitination, as these modifications can lead to different functional outcomes.
The literature contains some apparently contradictory findings regarding PPIL2's role in cancer, requiring careful methodological approaches to reconcile:
Researchers should avoid overgeneralizing PPIL2 function across cancer types and instead focus on defining context-specific mechanisms.
DNA repair pathway research is complex, and conflicting data regarding PPIL2's role may arise. To address such contradictions:
When addressing contradictory findings, researchers should carefully document experimental conditions, cell types, and damage induction methods to enable proper comparison across studies.
Cyclophilin-60 (Cyp60) is a member of the cyclophilin family of peptidyl-prolyl isomerases (PPIases), which are enzymes that catalyze the cis-trans isomerization of proline imidic peptide bonds in proteins. This family of enzymes is highly conserved and ubiquitous, playing crucial roles in various cellular processes, including protein folding, immunosuppression, and viral infection .
Cyclophilins, including Cyp60, are characterized by their ability to bind cyclosporin A (CsA), an immunosuppressive drug widely used in organ transplantation to prevent rejection. The binding of CsA to cyclophilins inhibits their PPIase activity, which is essential for their role in protein folding and function .
Cyp60, like other cyclophilins, has a conserved domain structure that includes a peptidyl-prolyl isomerase domain. This domain is responsible for the enzyme’s catalytic activity, facilitating the proper folding of proteins by accelerating the interconversion between cis and trans forms of proline residues .
Cyclophilins, including Cyp60, are involved in various biological processes. They act as chaperones, assisting in the proper folding of newly synthesized proteins and preventing the aggregation of misfolded proteins. Additionally, cyclophilins play a role in the immune response by modulating the activity of immune cells and influencing the production of cytokines .
Cyp60 has been implicated in several physiological and pathological processes. For instance, it has been shown to interact with HIV-1 virions, facilitating their replication and infection. This interaction makes cyclophilins potential targets for antiviral therapies .
Recombinant human cyclophilin-60 (rhCyp60) is produced using recombinant DNA technology, which involves the insertion of the human Cyp60 gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the protein for research and therapeutic purposes.
Recombinant cyclophilins, including rhCyp60, are valuable tools in biochemical and pharmacological studies. They are used to investigate the structure-function relationships of cyclophilins, their interactions with ligands such as CsA, and their roles in various cellular processes. Additionally, recombinant cyclophilins are employed in drug discovery efforts to develop isoform-selective inhibitors that can modulate cyclophilin activity for therapeutic benefit .
The study of cyclophilins, including Cyp60, has significant implications for understanding and treating various diseases. For example, cyclophilin inhibitors are being explored as potential therapies for viral infections, including HIV and hepatitis C, as well as for inflammatory and autoimmune diseases .
Furthermore, the development of isoform-specific cyclophilin inhibitors holds promise for targeted therapies with reduced side effects. By selectively inhibiting specific cyclophilin isoforms, researchers aim to achieve therapeutic benefits while minimizing the impact on other cellular processes .
In conclusion, Cyclophilin-60 (Human Recombinant) is a crucial member of the cyclophilin family with diverse roles in cellular processes and disease mechanisms. Its study and application in research and therapy continue to advance our understanding of protein folding, immune regulation, and potential therapeutic interventions.