PSME3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery time, please consult your local distributors.
Synonyms
11S regulator complex gamma subunit antibody; 11S regulator complex subunit gamma antibody; Activator of multicatalytic protease subunit 3 antibody; Ki antibody; Ki antigen antibody; Ki nuclear autoantigen antibody; Ki, PA28 gamma antibody; PA28 gamma antibody; PA28g antibody; PA28gamma antibody; Proteasome (prosome, macropain) activator subunit 3 (PA28 gamma, Ki) antibody; Proteasome (prosome, macropain) activator subunit 3 antibody; Proteasome activator 28 gamma antibody; Proteasome activator 28 subunit gamma antibody; Proteasome activator complex subunit 3 antibody; Proteasome activator subunit 3 antibody; PSME3 antibody; PSME3_HUMAN antibody; REG GAMMA antibody; REG-gamma antibody
Target Names
Uniprot No.

Target Background

Function
PSME3, also known as REG-gamma or PA28-gamma, is a subunit of the 11S REG-gamma proteasome regulator. This regulator forms a doughnut-shaped homoheptamer that associates with the proteasome. 11S REG-gamma activates the trypsin-like catalytic subunit of the proteasome while inhibiting the chymotrypsin-like and postglutamyl-preferring (PGPH) subunits. It facilitates the MDM2-p53/TP53 interaction, promoting ubiquitination- and MDM2-dependent proteasomal degradation of p53/TP53. This action limits p53/TP53 accumulation, resulting in inhibited apoptosis following DNA damage. PSME3 may also play a role in cell cycle regulation. It mediates CCAR2 and CHEK2-dependent SIRT1 inhibition.
Gene References Into Functions
  • Knockdown of REG-GAMMA (REGgamma) can inhibit the proliferation and migration, and promote the apoptosis of plasma cell myeloma RPMI-8226 cells, potentially by downregulating the NF-kappa-B (NF-kappaB) signaling pathway. PMID: 29020881
  • Research indicates that PIP30 significantly impacts PA28gamma interactions with cellular proteins, including the 20S proteasome. This suggests PIP30 acts as a crucial regulator of PA28gamma within cells, playing a critical role in controlling the diverse functions of the proteasome within the nucleus. PMID: 29934401
  • Studies have shown that the ubiquitin-independent REGgamma proteasome plays a role in regulating energy homeostasis. PMID: 27511885
  • High expression of REGg appears to be positively correlated with T-stage and lymph node metastasis in papillary thyroid carcinoma tissues. PMID: 29509725
  • Research demonstrates that REGgamma is a central molecule in the development of melanoma, regulating the Wnt/beta-catenin pathway. PMID: 28605165
  • Studies indicate that Proteasome activator subunit 3 induces epithelial-mesenchymal transition, leading to the expression of CSC markers and influencing the tumor immune microenvironment in breast cancer. PMID: 28529105
  • PSME3 plays an oncogenic role in pancreatic cancer by inhibiting c-Myc degradation, promoting glycolysis. PMID: 27756569
  • Gene therapy using proteasome activator, PA28gamma, has shown potential to improve ubiquitin-proteasome system function and alleviate behavioral abnormalities in Huntington's disease model mice. PMID: 26944602
  • PA28gamma overexpression has been identified as a potential surrogate prognostic biomarker in OSCC (Oral Squamous Cell Carcinoma). PMID: 26425675
  • Research has revealed significantly higher expression of PA28gamma in OSCC tumor tissues compared to normal tissues. PMID: 26425691
  • REGgamma plays a role in skin tumorigenesis by mediating MAPK/p38 activation of the Wnt/beta-catenin pathway. PMID: 25908095
  • Molecular cloning has identified a novel transcript variant encoding a truncated form of PA28G, potentially involved in cell cycle regulation and apoptosis. PMID: 25936920
  • In cellular contexts dominated by p53, pro-apoptotic signaling may be countered by PA28gamma-mediated caspase inhibition. PMID: 26201457
  • Studies suggest that high expression of REGgamma may predict metastasis and poor prognosis in breast cancer. PMID: 25550823
  • Elevated PA28gamma serum levels have been identified as a prognostic indicator of disease activity in rheumatoid arthritis. PMID: 25482151
  • Research indicates that gene expression levels of both PSME3 (proteasome activator subunit 3) and DUSP3 (dual specificity phosphatase 3) are associated with susceptibility to Staphylococcus aureus infection/sepsis in humans and mouse models. PMID: 24901344
  • Examination of EC and normal endometrium specimens confirmed the oncogenic role of REGgamma, revealing higher overexpression in p53-positive specimens compared to p53-negative specimens. PMID: 25697482
  • PA28 gamma and p53 form a negative feedback loop that maintains cellular balance of p53 and PA28gamma. PMID: 24531141
  • Data suggests that miR-7-5p plays a crucial role in inhibiting REGgamma in breast cancer cells. PMID: 25511742
  • REGgamma expression is positively correlated with ERalpha status and poor clinical prognosis in ERalpha-positive breast cancer patients. PMID: 25490392
  • Research links Chk2 and REGgamma to the mechanism underlying DBC1-dependent SIRT1 inhibition. PMID: 25361978
  • PKA turnover by the REGgamma-proteasome modulates FoxO1 cellular activity and VEGF-induced angiogenesis. PMID: 24560667
  • Expression of PA28gamma contributes to carcinogenesis and progression of colorectal cancer. PMID: 24113729
  • The REGgamma-proteasome pathway is differentially regulated by p53/TGF-beta signaling and mutant p53 in cancer cells. PMID: 24157709
  • Studies have shed light on the regulation of REGgamma assembly and activity, suggesting a potential avenue for intervention in ubiquitin-independent REGgamma proteasome activity. PMID: 23612972
  • PA28gamma functions as a co-repressor of HTLV-1 p30, suppressing virus replication and contributing to the maintenance of viral latency. PMID: 23104922
  • Statistical analysis of laryngeal carcinomas revealed a positive relationship between the levels of REGgamma and the expression of p53 and p2. This suggests that REGgamma overexpression may facilitate the growth of laryngeal cancer cells. PMID: 22938444
  • PA28gamma is an ATM target, being recruited to DNA damage sites where it is essential for the rapid accumulation of proteasomes and the timely coordination of DNA double-strand break repair. PMID: 22134242
  • REG-gamma associates with and modulates the abundance of nuclear activation-induced deaminase. PMID: 22042974
  • Research has uncovered a previously unrecognized mechanism regulating the activity of the proteasome activator REGgamma. PMID: 21445096
  • High REGgamma expression is linked to breast cancer and its metastatic lymph nodes. PMID: 20467919
  • HTLV-1 p30 interacts with ATM and REGgamma, promoting viral spread by facilitating cell survival. PMID: 21216954
  • REGgamma regulates cellular distribution of p53 by facilitating its multiple monoubiquitylation and nuclear export. PMID: 21084564
  • REGgamma-mediated p53 proteolysis contributes to the proviral function of REGgamma. The host REGgamma pathway is utilized and modified during CVB3 infection, promoting efficient viral replication. PMID: 20719955
  • REGgamma is widely expressed in many tissues, with the highest expression observed in the testis. PMID: 20494959
  • PA28gamma participates in both the pathogenesis and propagation of HCV by regulating the degradation of the core protein in both a ubiquitin-dependent and ubiquitin-independent manner. PMID: 20683941
  • Overexpression of the proteasome activator subunit PA28gamma restored proteasome function in Huntington disease cells. This improved cell viability in mutant huntingtin-expressing striatal neurons exposed to pathological stressors. PMID: 17327906
  • Research suggests that REGgamma promotes tumor growth through complex multi-factor mechanisms. PMID: 19656465
  • PA28gamma is an endogenous substrate for caspase-3 and -7. PMID: 11859414
  • Ki antigen contains multiple epitopes recognized by autoimmune sera. PMID: 12784391
  • PA28 gamma acts as a novel regulator of Cajal body integrity in response to ultraviolet radiation. PMID: 17088425
  • REGgamma proteasome activator is involved in maintaining chromosomal stability. PMID: 18235248
  • PA28gamma, a proteasome activator that inhibits apoptosis and promotes cell cycle progression through unknown mechanisms, acts as a cofactor in the MDM2-p53 interaction. PMID: 18309296
  • PA28gamma/REGgamma, which specifically binds to hepatitis c virus core protein, is required for the virulence of the core protein. PMID: 18321762
  • Mammalian proteasomes are unable to degrade glutamine-expanded regions within pathogenic polyQ-expanded proteins, such as Huntingtin. PMID: 18343811
  • PA28gamma specifically binds to hepatitis C virus core protein and is involved in its degradation. PMID: 19091860
  • Research highlights a new role for REGgamma in the control and regulation of promyelocytic leukemia subnuclear structures. PMID: 19556897

Show More

Hide All

Database Links

HGNC: 9570

OMIM: 605129

KEGG: hsa:10197

STRING: 9606.ENSP00000293362

UniGene: Hs.152978

Protein Families
PA28 family
Subcellular Location
Nucleus. Cytoplasm.

Q&A

What is PSME3 and what is its biological significance?

PSME3 (Proteasome activator subunit 3) is a critical component of the 11S REG-gamma (also called PA28-gamma) proteasome regulator. It forms a doughnut-shaped homoheptamer that associates with the proteasome and selectively activates the trypsin-like catalytic subunit while inhibiting the chymotrypsin-like and postglutamyl-preferring subunits . Located on chromosome 17q21.31, PSME3 has multiple biological roles including:

  • Facilitating MDM2-p53/TP53 interaction that promotes ubiquitination and proteasomal degradation of p53/TP53

  • Regulating the degradation of cell cycle inhibitor p21

  • Promoting the degradation of SRC-3 proteins through ubiquitin- and ATP-independent mechanisms

  • Participating in cell cycle regulation

  • Mediating CCAR2 and CHEK2-dependent SIRT1 inhibition

Recent research has revealed PSME3's significant involvement in various cancers and immune regulation processes, making it a promising target for cancer research and potential therapeutic development .

What are the primary research applications for PSME3 antibodies?

PSME3 antibodies serve as essential tools for investigating this protein's expression, localization, and function across multiple experimental contexts:

  • Western Blotting (WB): For quantitative assessment of PSME3 protein levels in cell and tissue lysates

  • Immunoprecipitation (IP): To study protein-protein interactions involving PSME3

  • Immunohistochemistry-Paraffin (IHC-P): For examining PSME3 distribution and expression in tissue sections

  • Flow Cytometry: For analyzing PSME3 in relation to other cellular markers, particularly in immune cells

  • Immunofluorescence: For subcellular localization studies

These applications have been instrumental in establishing PSME3's roles in cancer development, immune regulation, and its potential as a prognostic biomarker .

How should researchers select appropriate PSME3 antibodies for their experiments?

Selection of PSME3 antibodies should be based on several critical factors:

  • Experimental application: Different applications (WB, IHC-P, IP, etc.) may require antibodies with specific properties

  • Species reactivity: Confirm the antibody recognizes PSME3 in your experimental model (human, mouse, etc.)

  • Epitope location: Consider whether the epitope is within a functional domain that might be masked in certain contexts

  • Validation data: Review existing validation in literature and manufacturer data

  • Clonality: Polyclonal antibodies (like ab157157) offer high sensitivity and recognize multiple epitopes, while monoclonal antibodies provide higher specificity for a single epitope

When studying complex interactions or specific domains of PSME3, consider the immunogen information. For example, ab157157 was raised against a synthetic peptide within the C-terminal region (aa 200 to C-terminus) of human PSME3 .

What are the optimal protocols for using PSME3 antibodies in Western blotting?

Western blotting with PSME3 antibodies requires careful optimization:

  • Sample preparation:

    • Use RIPA or NP-40 buffer supplemented with protease inhibitors

    • Include phosphatase inhibitors if studying phosphorylation status

    • Maintain samples at 4°C during lysis to prevent degradation

  • Gel electrophoresis:

    • 10-12% SDS-PAGE gels are typically suitable for resolving PSME3 (~30 kDa)

    • Load appropriate positive controls (e.g., cell lines known to express PSME3)

  • Transfer and detection:

    • PVDF membranes often provide better results than nitrocellulose

    • Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature

    • Incubate with primary PSME3 antibody (typically 1:1000 dilution) overnight at 4°C

    • Use appropriate HRP-conjugated secondary antibody (typically 1:5000-1:10000)

    • Develop using ECL reagents optimized for the expected expression level

  • Controls:

    • Include PSME3 knockdown/overexpression samples as specificity controls

    • Consider GAPDH, β-actin, or α-tubulin as loading controls

For cancer studies, A549 (lung adenocarcinoma), Hut7 (liver cancer), and T24 (bladder cancer) cell lines have been validated for PSME3 expression and can serve as positive controls .

How can researchers effectively use PSME3 antibodies for immunohistochemistry?

For optimal immunohistochemistry results with PSME3 antibodies:

  • Tissue preparation:

    • Fix tissues in 10% neutral buffered formalin for 24-48 hours

    • Process and embed in paraffin following standard protocols

    • Cut sections at 4-5 μm thickness

  • Antigen retrieval:

    • Heat-induced epitope retrieval in citrate buffer (pH 6.0) is typically effective

    • Pressure cooker methods often yield superior results

  • Staining protocol:

    • Block endogenous peroxidase activity with 3% H₂O₂

    • Block non-specific binding with serum-free protein block

    • Incubate with PSME3 primary antibody (1:100-1:200 dilution) overnight at 4°C

    • Use appropriate detection system (HRP-polymer or ABC method)

    • Counterstain with hematoxylin, dehydrate, and mount

  • Controls and scoring:

    • Include positive control tissues based on The Human Protein Atlas data

    • Implement negative controls (primary antibody omission)

    • Score based on intensity (0-3) and percentage of positive cells

    • Consider both nuclear and cytoplasmic staining as PSME3 functions in both compartments

This approach has been validated in studies examining PSME3 expression across multiple cancer types, confirming high consistency between protein levels detected by IHC and mRNA expression data .

What approaches should be used for PSME3 knockdown or overexpression validation studies?

When conducting PSME3 manipulation studies, proper validation is essential:

  • For PSME3 overexpression:

    • Use pCDNA3.1-Flag-PSME3 plasmid for transfection-based approaches

    • For viral delivery, use an optimal ratio of pMD2G:psPAX2:plasmid:PEI = 1:2:4:12 (μg:μg:μg:μl) for packaging in HEK 293T cells

    • Confirm overexpression by Western blot, qRT-PCR, and immunofluorescence

  • For PSME3 knockdown:

    • Design multiple siRNA/shRNA sequences targeting different regions

    • When using lentiviral vectors, infect cells at approximately 40% density

    • Change medium on day 2 post-infection

    • Begin selection with appropriate antibiotics on day 3

    • Validate knockdown efficiency using multiple methods (Western blot, qRT-PCR)

  • Functional validation approaches:

    • Cell proliferation assays (CCK-8) at multiple time points (24h, 48h)

    • Migration/invasion assays (wound healing, transwell)

    • Cell cycle analysis by flow cytometry

    • Apoptosis assessment (Annexin V/PI staining)

These validation approaches have been successfully employed in studies demonstrating PSME3's role in promoting lung adenocarcinoma cell proliferation, migration, invasion, and inhibiting apoptosis .

How can PSME3 antibodies be used to investigate immune regulation mechanisms?

PSME3 plays significant roles in immune regulation that can be investigated using specialized antibody-based approaches:

  • Immune checkpoint analysis:

    • Use flow cytometry with dual staining for PSME3 and immune checkpoint proteins

    • Prepare antibody combinations (1:100 dilution) including anti-PSME3 and anti-CD276 (B7-H3)

    • Stain resuspended cells on ice for 30 minutes

    • Wash twice and fix with 1% paraformaldehyde

    • Analyze using flow cytometry software (e.g., BD Diva, FlowJo)

  • Tumor microenvironment studies:

    • Multiplex immunofluorescence combining PSME3 with markers for:

      • Neutrophils (CD15+)

      • B cells (CD19+, CD20+)

      • T cells (CD4+, CD8+)

      • M2 macrophages (CD68+, CD163+)

    • Analysis of spatial co-localization patterns

    • Correlation with immunoScore, stromalScore, and ESTIMATEScore

  • Immune regulation mechanisms:

    • Co-immunoprecipitation to identify PSME3 interactions with immune regulators

    • ChIP assays to examine transcriptional regulation of immune-related genes

    • Proximity ligation assays to confirm direct protein interactions in situ

This approach has revealed PSME3's association with immune checkpoints and confirmed its role in positively regulating CD276 expression, suggesting PSME3 as a potential therapeutic target in immunotherapy .

What methodological considerations are important when studying PSME3 in different cancer models?

When investigating PSME3 across different cancer models, researchers should consider several methodological aspects:

This comprehensive approach reveals cancer-specific roles of PSME3, as demonstrated by studies showing its correlation with adverse clinical outcomes and cancer progression particularly in liver cancer (LIHC) .

How should researchers interpret conflicting PSME3 expression data across different experimental platforms?

Researchers often encounter discrepancies in PSME3 expression data between different experimental platforms. A systematic approach to resolving these conflicts includes:

  • Platform-specific considerations:

    • RNA-seq vs. qRT-PCR: RNA-seq measures total transcript abundance while qRT-PCR may be isoform-specific

    • Protein detection methods: Western blot quantifies total protein, while IHC reveals spatial distribution

    • Flow cytometry: Measures per-cell expression but may be affected by fixation conditions

  • Analytical approach for reconciling discrepancies:

    • Begin with biological replicates to establish consistency within each platform

    • Perform parallel validation using orthogonal methods

    • Consider splice variants and post-translational modifications

    • Evaluate antibody epitope accessibility in different experimental conditions

    • Assess cell-type specific expression in heterogeneous samples

  • Methodology for comprehensive validation:

    • Employ multiple antibodies targeting different epitopes

    • Include positive controls with known PSME3 expression

    • Validate with genetic approaches (knockdown/overexpression)

    • Consider the impact of tumor heterogeneity in cancer studies

Research demonstrates that while PSME3 mRNA and protein levels generally show high consistency across cancer types, microenvironmental factors can influence expression patterns and protein function in ways not reflected at the transcript level .

How can PSME3 antibodies contribute to biomarker development in cancer immunotherapy?

PSME3 shows promise as a biomarker for cancer immunotherapy response, with antibody-based detection playing a central role:

  • Biomarker potential assessment methodology:

    • Correlate PSME3 expression with established immunotherapy markers:

      • TMB (Tumor Mutational Burden)

      • MSI (Microsatellite Instability)

      • MMR gene expression

    • Analyze relationship with immune checkpoint expression

    • Evaluate association with immune cell infiltration patterns

  • Implementation strategies:

    • Develop standardized IHC protocols for clinical sample analysis

    • Establish scoring systems that account for intensity and distribution

    • Create multiplex assays combining PSME3 with other immune markers

    • Validate cut-off values for stratifying patient responses

  • Clinical validation approach:

    • Retrospective analysis correlating PSME3 expression with treatment outcomes

    • Prospective studies in patients receiving immune checkpoint inhibitors

    • Investigation of PSME3 as a companion diagnostic for novel immunotherapies

Research has shown that PSME3 positively regulates CD276 (B7-H3) expression, suggesting its potential relevance as a biomarker for immunotherapy response. The association between PSME3 and TMB/MSI status further supports its utility in predicting response to immune checkpoint inhibitors across multiple cancer types .

What are the cutting-edge approaches for studying PSME3 interactions with the proteasome system?

Advanced techniques for investigating PSME3-proteasome interactions include:

  • Structural biology approaches:

    • Cryo-EM analysis of PSME3-proteasome complexes

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • FRET-based assays to monitor dynamic associations in living cells

  • Proteasomal activity assessment:

    • Fluorogenic substrate assays measuring trypsin-like, chymotrypsin-like, and PGPH activities

    • Cell-based proteasome sensors to monitor activity in real-time

    • In vitro reconstitution assays with purified components

  • Protein interaction network mapping:

    • Proximity labeling approaches (BioID, APEX) to identify transient interactions

    • Quantitative interaction proteomics following immunoprecipitation

    • Yeast two-hybrid or mammalian two-hybrid screens for novel interactors

Research has identified key PSME3-interacting proteins including PSME1, PSME2, PSMA5, PSMD8, PSMD14, PSMEIP1, NCOA3, RXRA, TNF, and IFNG. Additionally, the interaction between PSME3 and PIP30 enhances its binding to the cellular 20S proteasome and affects its substrate specificity .

How can single-cell analysis techniques be integrated with PSME3 antibody applications?

Single-cell analysis offers unprecedented insights into PSME3 biology at the cellular level:

  • Single-cell proteomics approaches:

    • Mass cytometry (CyTOF) with metal-conjugated PSME3 antibodies

    • CITE-seq combining PSME3 antibodies with transcriptome analysis

    • Imaging mass cytometry for spatial resolution of PSME3 expression

  • Spatial transcriptomics integration:

    • Correlate PSME3 spatial expression with immune cell markers

    • Validate co-expression patterns observed in spatial transcriptome data

    • Map PSME3 expression to specific tissue microenvironments

  • Single-cell functional assays:

    • Microfluidic approaches for correlating PSME3 levels with cellular phenotypes

    • Live-cell imaging with fluorescently tagged antibodies or nanobodies

    • Single-cell secretome analysis in relation to PSME3 expression

These approaches have revealed that PSME3 exhibits spatial co-expression with M2 macrophage biomarkers (CD68 and CD163) in certain tissues, with overlapping geographical distribution patterns that suggest functional relationships in the tumor microenvironment .

How can researchers address common challenges when working with PSME3 antibodies?

When working with PSME3 antibodies, researchers may encounter several challenges that require systematic troubleshooting:

  • Western blot issues:

    • Non-specific bands: Optimize antibody dilution (typically 1:1000), increase blocking time, and use PVDF membranes

    • Weak signal: Increase protein loading, extend primary antibody incubation time, or use enhanced detection systems

    • High background: Increase washing steps, reduce secondary antibody concentration, and use freshly prepared buffers

  • Immunohistochemistry challenges:

    • Variable staining: Standardize fixation time, optimize antigen retrieval (citrate buffer, pH 6.0), and titrate antibody

    • Background staining: Increase blocking time, use avidin/biotin blocking for biotin-based detection systems

    • Loss of antigenicity: Minimize storage time of cut sections and use freshly prepared buffers

  • Flow cytometry considerations:

    • Fixation effects: Compare results with different fixation methods (1% paraformaldehyde is recommended)

    • Antibody penetration: Include permeabilization step when analyzing intracellular PSME3

    • Compensation: Properly compensate when combining PSME3 detection with other fluorochromes

  • Validation approaches:

    • Always include positive controls (A549, Hut7, T24 cell lines)

    • Perform siRNA knockdown as negative control

    • Consider testing multiple antibody clones when results are inconsistent

These troubleshooting approaches are essential for generating reliable and reproducible data when studying PSME3 in various experimental contexts.

What are the best practices for quantitative analysis of PSME3 expression?

Accurate quantification of PSME3 expression requires rigorous methodology:

  • Western blot quantification:

    • Use linear range of detection (avoid saturated signals)

    • Normalize to appropriate loading controls (GAPDH, β-actin, total protein)

    • Apply densitometry using software like ImageJ with background subtraction

    • Include calibration standards when absolute quantification is needed

  • Immunohistochemistry scoring:

    • Implement standardized scoring systems (H-score = intensity × percentage)

    • Intensity scale: 0 (negative), 1+ (weak), 2+ (moderate), 3+ (strong)

    • Percentage of positive cells: 0-100%

    • Consider automated image analysis for reproducibility

    • Account for both nuclear and cytoplasmic staining independently

  • qRT-PCR quantification:

    • Use validated reference genes (GAPDH, ACTB, 18S rRNA)

    • Apply delta-delta Ct (2^-ΔΔCt) method for relative quantification

    • Include standard curves for absolute quantification

    • Account for PCR efficiency in calculations

  • Flow cytometry analysis:

    • Report mean fluorescence intensity (MFI) and percentage of positive cells

    • Use isotype controls to set negative thresholds

    • Apply consistent gating strategies across experiments

These quantitative approaches have been instrumental in establishing the prognostic significance of PSME3 expression across various cancer types, particularly in lung adenocarcinoma and liver cancer .

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.