FKBP3 Human

FK506 Binding Protein 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of FKBP3 Human

FKBP3 (FK506-binding protein 3) is a 25.1 kDa cis-trans prolyl isomerase belonging to the immunophilin protein family. It plays critical roles in protein folding, immunoregulation, and cellular signaling. Structurally, FKBP3 consists of 224 amino acids and is non-glycosylated, with a high affinity for the immunosuppressant rapamycin compared to FK506 .

PropertyDetails
Molecular Weight25.1 kDa
Amino Acids224
Expression SystemE. coli (recombinant)
Key LigandsRapamycin, FK506, HDAC1/2, YY1, casein kinase II, nucleolin

Key Functional Roles

  1. Immunosuppression: Binds rapamycin to inhibit mTORC1/2 complexes, affecting T-cell proliferation and metabolism .

  2. Epigenetic Regulation: Associates with histone deacetylases (HDAC1/2) to repress transcriptional activation of cell cycle inhibitors like p27 .

  3. Viral Latency: Indirectly maintains HIV-1 latency by modulating chromatin accessibility .

Protein-Protein Interactions

PartnerFunctionReference
MTORRegulates mTORC1/2 signaling complexes
HDAC1/2Enhances histone deacetylation to repress tumor suppressor genes
YY1Modulates transcriptional repression via chromatin remodeling
Casein Kinase IIPhosphorylates transcription factors to regulate gene expression
NucleolinInvolvement in RNA processing and ribosome biogenesis

HIV-1 Latency

FKBP3 knockout in latently infected cells reduces viral persistence, suggesting its role in maintaining chromatin silencing. This was demonstrated in CD4+ T lymphocytes, where FKBP3 depletion increased viral reactivation without significant cell activation .

Non-Small Cell Lung Cancer (NSCLC)

FKBP3 promotes proliferation by suppressing p27 expression via HDAC2-mediated epigenetic silencing. Clinical studies show high FKBP3 expression correlates with poor prognosis and lymph node metastasis .

StudyMechanismOutcome
FKBP3 knockdown (NSCLC)↓ HDAC2, ↑ p27 → G1/S cell cycle arrestSlowed tumor growth
miR-145-5p targeting↓ FKBP3 → ↓ Sp1/HDAC2 signalingReduced in vivo tumor size

Colorectal Cancer (CRC) Drug Resistance

FKBP3 mediates oxaliplatin resistance by upregulating P-gp and activating the PTEN/AKT pathway. Knockdown restores drug sensitivity and induces apoptosis .

Clinical and Therapeutic Implications

  • Oncology: FKBP3’s role in HDAC2 regulation makes it a potential target for combining epigenetic inhibitors with conventional therapies in NSCLC and CRC .

  • Immunology: Its interaction with mTOR highlights therapeutic potential in autoimmune diseases, though rapamycin’s broader effects complicate specificity .

  • Virology: Targeting FKBP3 in HIV-1 latency reversal strategies could enhance antiretroviral therapies .

Product Specs

Introduction
FKBP3, a member of the immunosuppressant receptor family, plays a crucial role in inhibiting T-cell proliferation. It achieves this by halting two distinct cytoplasmic signal transmission pathways. Functioning as a protein folding facilitator, FKBP3 belongs to the immunophilin protein family, which is involved in immunoregulation and essential cellular processes such as protein folding and trafficking. This cis-trans prolyl isomerase enzyme exhibits binding affinity for immunosuppressants like FK506 and rapamycin, in addition to histone deacetylases, the transcription factor YY1, casein kinase II, and nucleolin. Notably, its affinity for rapamycin surpasses that of FK506, highlighting its significance in rapamycin-mediated immunosuppression.
Description
Produced in E.Coli, FKBP3 is a single, non-glycosylated polypeptide chain consisting of 224 amino acids (amino acids 1-224). Its molecular weight is 25.1 kDa. The purification of FKBP3 is carried out using proprietary chromatographic techniques.
Physical Appearance
A colorless solution that has been sterilized by filtration.
Formulation
The FKBP3 protein solution is formulated with 20mM Tris-HCl at a pH of 8, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 90.0%.
Biological Activity

The specific activity, determined by measuring the enzyme's ability to cleave 1 µmole of suc-AAFP-pNA per minute at 37°C in Tris-Hcl pH 8.0 using chymotrypsin as a reference, is greater than 900 nmoles/min/mg.

Synonyms
EC 5.2.1.8, FKBP-3, FKBP25, FKBP-25, Peptidyl-prolyl cis-trans isomerase FKBP3, PPIase FKBP3, FK506-binding protein 3, Rotamase, Immunophilin FKBP25, 25 kDa FK506-binding protein, 25 kDa FKBP, Rapamycin-selective 25 kDa immunophilin, FKBP3, PPIase.
Source
Escherichia Coli.
Amino Acid Sequence
MAAAVPQRAW TVEQLRSEQL PKKDIIKFLQ EHGSDSFLAE HKLLGNIKNV AKTANKDHLV TAYNHLFETK RFKGTESISK VSEQVKNVKL NEDKPKETKS EETLDEGPPK YTKSVLKKGD KTNFPKKGDV VHCWYTGTLQ DGTVFDTNIQ TSAKKKKNAK PLSFKVGVGK VIRGWDEALL TMSKGEKARL EIEPEWAYGK KGQPDAKIPP NAKLTFEVEL VDID.

Q&A

What is FKBP3 and what is its primary function in human cells?

FKBP3, also known as FKBP25, is a protein encoded by the FKBP3 gene in humans. It belongs to the immunophilin protein family, which plays crucial roles in immunoregulation and basic cellular processes involving protein folding and trafficking. FKBP3 functions as a cis-trans prolyl isomerase that binds the immunosuppressants FK506 and rapamycin, with significantly higher affinity for rapamycin than for FK506 . This binding property suggests that FKBP3 may serve as an important target molecule for immunosuppression by rapamycin.

The protein is involved in multiple cellular functions beyond immunoregulation, including:

  • Protein folding and trafficking

  • Epigenetic regulation through interactions with histone deacetylases

  • Transcriptional regulation through interaction with transcription factors

  • Potential roles in DNA damage response pathways

The multifaceted nature of FKBP3 has made it an interesting target for various research fields, from immunology to virology and cancer biology .

How is FKBP3 structurally characterized and what protein domains does it contain?

FKBP3 has been structurally characterized through both crystallography and NMR studies. The crystal structure of FKBP25 (FKBP3) with FK506 has been published with PDB ID 5D75, while the NMR structure of full-length FKBP25 is available with PDB ID 2MPH .

The protein contains:

  • A PPIase (peptidyl-prolyl cis-trans isomerase) domain that catalyzes the isomerization of proline residues in peptide bonds

  • Regions that facilitate interactions with other proteins, including YY1, HDAC1/2, and DNA

  • Structural elements that enable binding to immunosuppressive drugs like FK506 and rapamycin

These structural characteristics enable FKBP3 to function in various cellular contexts, including protein folding, immunoregulation, and epigenetic modification through its interactions with histone deacetylases .

What key protein-protein interactions have been identified for FKBP3?

FKBP3 engages in several critical protein-protein interactions that underlie its diverse cellular functions:

  • YY1 (Yin Yang 1): FKBP3 interacts with this transcription factor, potentially influencing gene expression regulation. This interaction is particularly important in the context of HIV-1 latency .

  • HDAC1/2 (Histone Deacetylases 1 and 2): FKBP3 associates with these histone-modifying enzymes, suggesting a role in epigenetic regulation. Co-immunoprecipitation assays have confirmed these interactions in HIV latency cell models .

  • DNA binding: FKBP3 has been shown to interact directly with DNA, which may contribute to its role in transcriptional regulation and chromatin organization .

  • Mdm2: This interaction suggests involvement in p53 regulation pathways and potentially cell cycle control .

  • PARK7 (DJ-1): In diffuse large B-cell lymphoma (DLBCL) cells, FKBP3 has been shown to interact with PARK7, contributing to the activation of the Wnt/β-catenin signaling pathway .

These interactions highlight FKBP3's role as a multifunctional adaptor protein that bridges various cellular processes including gene expression, epigenetic regulation, and signaling pathways .

How does FKBP3 contribute to HIV-1 latency maintenance?

FKBP3 plays a crucial role in establishing and maintaining HIV-1 latency through an epigenetic regulation mechanism. Research has revealed a specific molecular pathway:

  • FKBP3 serves as an adaptor protein that interacts with the transcription factor YY1, which can bind to the HIV-1 long terminal repeat (LTR) region .

  • Through this interaction, FKBP3 indirectly associates with the HIV-1 LTR. Chromatin immunoprecipitation (ChIP) and quantitative PCR experiments have confirmed this association .

  • FKBP3 subsequently recruits histone deacetylases (HDAC1/2) to the HIV-1 LTR region .

  • The recruited HDAC1/2 promotes histone deacetylation at the HIV-1 promoter region, which leads to chromatin compaction and transcriptional silencing .

  • This epigenetic modification creates and maintains a repressive chromatin environment at the HIV-1 promoter, thereby contributing to viral latency .

This mechanism highlights FKBP3's role as a scaffold protein that facilitates the recruitment of epigenetic modifiers to the HIV-1 promoter, ultimately promoting viral latency through chromatin remodeling .

What experimental approaches are used to study FKBP3's role in HIV-1 latency?

Researchers employ multiple experimental approaches to investigate FKBP3's role in HIV-1 latency:

  • CRISPR/Cas9 Gene Knockout:

    • Guide RNAs (sgRNAs) targeting FKBP3 are designed and delivered via lentiviral vectors

    • Puromycin selection (typically 2 μg/ml for 14 days) is used to isolate knockout cells

    • Genomic DNA sequencing confirms successful edits at target sites with various indels

    • Western blotting validates FKBP3 protein depletion

  • Latency Model Cell Lines:

    • Multiple latently infected cell models are utilized, including:

      • C11 cells (containing HIV-1 proviral DNA with GFP reporter)

      • J-Lat 10.6 cells

      • ACH2 cells

    • Flow cytometry analysis measures GFP expression as an indicator of HIV-1 reactivation

  • Protein-Protein Interaction Studies:

    • Co-immunoprecipitation (Co-IP) assays confirm interactions between FKBP3, YY1, and HDAC1/2

    • These assays typically use antibodies against FKBP3 followed by Western blotting for interacting partners

  • Chromatin Association Analysis:

    • Chromatin immunoprecipitation (ChIP) followed by qPCR determines FKBP3 binding to the HIV-1 LTR

    • This confirms direct association with viral genomic elements

  • Primary Cell Models:

    • Primary CD4+ T lymphocytes from multiple donors are used to create physiologically relevant latency models

    • NanoLuc luciferase reporters monitor HIV-1 expression levels

    • Electroporation delivers CRISPR components for FKBP3 knockout in primary cells

These methodologies collectively provide robust evidence for FKBP3's mechanistic role in HIV-1 latency establishment and maintenance .

What is the effect of FKBP3 knockout on latent HIV-1 activation in different cell models?

FKBP3 knockout consistently activates latent HIV-1 across multiple cellular models, although with varying efficacy:

  • C11 Cell Model:

    • FKBP3 knockout induced reactivation of latent HIV-1 by approximately 30%

    • This effect was statistically significant and reproducible across experiments

    • GFP reporter expression was used to quantify viral reactivation

  • J-Lat 10.6 Cell Line:

    • Similar reactivation patterns were observed in this widely used latency model

    • The percent of GFP-positive cells increased significantly following FKBP3 knockout

    • This model further validated the findings from the C11 cells

  • ACH2 Cell Line:

    • FKBP3 knockout also effectively activated latent HIV-1 in this T-cell line

    • The consistency across different cell models strengthens the evidence for FKBP3's role in latency

  • Primary CD4+ T Lymphocyte Model:

    • Most significantly, FKBP3 knockout activated latent HIV-1 in primary CD4+ T cells

    • The primary cell model involved gradual decrease in IL-2 levels to establish latency

    • NanoLuc luciferase expression was measured to monitor HIV-1 activation

    • Importantly, FKBP3 expression levels increased significantly upon HIV-1 infection in primary CD4+ T cells, suggesting involvement in the immune response to infection

Control experiments confirmed that the observed effects were specifically due to FKBP3 knockout rather than cellular activation or apoptosis induced by experimental procedures. Notably, FKBP3 knockout did not significantly affect cell proliferation or apoptosis, suggesting its potential as a therapeutic target .

How does FKBP3 influence the progression of diffuse large B-cell lymphoma (DLBCL)?

FKBP3 plays a significant role in promoting the malignant phenotype of diffuse large B-cell lymphoma (DLBCL) through multiple mechanisms:

  • Enhanced Cellular Proliferation:

    • FKBP3 has been demonstrated to significantly increase the proliferation rate of DLBCL cells

    • This effect contributes to faster tumor growth and progression

  • Promotion of Cancer Stemness:

    • FKBP3 enhances stemness characteristics in DLBCL cells

    • Cancer stem cells are associated with therapy resistance and tumor recurrence

    • This suggests FKBP3 may contribute to treatment failures and disease relapse

  • Accelerated Tumor Growth in vivo:

    • Xenograft mouse models showed that FKBP3 aggravates tumor growth

    • This confirms the in vitro findings and establishes in vivo relevance

  • Molecular Pathway Activation:

    • FKBP3 activates the Wnt/β-catenin signaling pathway in DLBCL

    • This activation is mediated through PARK7 (DJ-1)

    • The Wnt/β-catenin pathway is known to drive proliferation and stemness in various cancers

  • Protein Stabilization Mechanism:

    • FKBP3 prevents the degradation of PARK7 by reducing its ubiquitination

    • Knockdown of FKBP3 enhances PARK7 degradation through increased ubiquitination modification

    • This establishes FKBP3 as a post-translational regulator of oncogenic proteins

These findings collectively establish FKBP3 as a potential therapeutic target for DLBCL treatment, as it contributes to multiple aspects of lymphoma pathogenesis .

What is the relationship between FKBP3 and FOXO3 in regulating DLBCL progression?

The relationship between FKBP3 and FOXO3 in diffuse large B-cell lymphoma (DLBCL) represents a critical regulatory axis:

  • Transcriptional Repression:

    • FOXO3 (Forkhead Box O3), a transcription factor from the forkhead family, directly binds to the promoter region of the FKBP3 gene

    • This binding leads to suppression of FKBP3 transcription

    • Consequently, FOXO3 negatively regulates FKBP3 expression at the transcriptional level

  • Tumor Suppressive Function:

    • FOXO3 acts as a tumor suppressor in DLBCL

    • By inhibiting FKBP3 expression, FOXO3 weakens the progression of DLBCL

    • This establishes a mechanism through which FOXO3 exerts its anti-cancer effects

  • Regulatory Pathway:

    • The FOXO3-FKBP3-PARK7-Wnt/β-catenin signaling axis forms a regulatory pathway in DLBCL

    • FOXO3 suppresses FKBP3, which normally stabilizes PARK7

    • Reduced PARK7 leads to decreased activation of the Wnt/β-catenin pathway

    • This ultimately results in diminished malignant characteristics of DLBCL cells

  • Therapeutic Implications:

    • The FOXO3-FKBP3 relationship suggests potential therapeutic strategies:

      • Enhancing FOXO3 activity could downregulate FKBP3 expression

      • This would mimic the effects of FKBP3 knockdown or inhibition

      • Such approaches could potentially suppress DLBCL progression

This regulatory relationship provides valuable insights into the molecular mechanisms underlying DLBCL pathogenesis and identifies potential points of therapeutic intervention in the FOXO3-FKBP3-PARK7-Wnt/β-catenin axis .

What are the recommended techniques for studying FKBP3 protein-protein interactions?

Several specialized techniques are recommended for investigating FKBP3 protein-protein interactions, each with specific advantages:

  • Co-Immunoprecipitation (Co-IP):

    • Most commonly used for FKBP3 interaction studies

    • Protocol:

      • Lyse cells in appropriate buffer (typically containing protease inhibitors)

      • Incubate lysates with anti-FKBP3 antibody (or antibodies against suspected interacting partners)

      • Capture complexes with protein A/G beads

      • Wash extensively to remove non-specific interactions

      • Analyze precipitated proteins by Western blotting

    • Successfully demonstrated interactions between FKBP3 and YY1, HDAC1/2 in HIV latency models

  • Chromatin Immunoprecipitation (ChIP):

    • Essential for studying FKBP3 interactions with DNA and chromatin

    • Protocol highlights:

      • Crosslink protein-DNA complexes with formaldehyde

      • Sonicate chromatin to appropriate fragment size (200-500bp)

      • Immunoprecipitate with anti-FKBP3 antibodies

      • Analyze enriched DNA regions by qPCR or sequencing

    • Has confirmed FKBP3 binding to HIV-1 LTR regions

  • Proximity Ligation Assay (PLA):

    • Detects protein interactions in situ with high sensitivity

    • Particularly useful for visualizing FKBP3 interactions within cellular compartments

    • Can confirm interactions identified by co-IP in their native cellular context

  • Pull-down Assays with Recombinant Proteins:

    • Useful for determining direct versus indirect interactions

    • Protocol approach:

      • Express and purify tagged recombinant FKBP3

      • Incubate with cell lysates or purified candidate proteins

      • Capture complexes via the tag

      • Identify interacting partners by mass spectrometry or Western blotting

  • FRET (Förster Resonance Energy Transfer) or BiFC (Bimolecular Fluorescence Complementation):

    • Enables visualization of protein interactions in living cells

    • Can provide spatial and temporal information about FKBP3 interactions

When studying FKBP3 interactions, researchers should consider potential confounding factors such as the presence of immunosuppressive drugs (FK506, rapamycin) which may alter interaction profiles, and the cellular compartmentalization of FKBP3, which can vary depending on cellular conditions .

What are effective methods for modulating FKBP3 expression in experimental systems?

Researchers employ several complementary approaches to effectively modulate FKBP3 expression in experimental systems:

  • CRISPR/Cas9-mediated Gene Knockout:

    • Most definitive approach for complete elimination of FKBP3

    • Implementation details:

      • Design multiple sgRNAs targeting early exons of FKBP3 (typically 3 different guides)

      • Deliver via lentiviral vectors with appropriate selection markers

      • Select with puromycin (typically 2 μg/ml for ~14 days)

      • Confirm knockout by genomic DNA sequencing and Western blotting

    • Successfully used in multiple cell models including C11, J-Lat 10.6, and ACH2 cells

  • RNA Interference (RNAi):

    • Provides more rapid and often reversible knockdown

    • Implementation approaches:

      • siRNA for transient knockdown (72-96 hours)

      • shRNA via lentiviral vectors for stable knockdown

      • Typical knockdown efficiency: 70-90% reduction in FKBP3 protein levels

    • Useful for dose-response studies by varying knockdown efficiency

  • Inducible Expression Systems:

    • For controlled overexpression or knockdown

    • Options include:

      • Tetracycline-regulated systems (Tet-On/Tet-Off)

      • Allows temporal control of FKBP3 expression

      • Useful for studying time-dependent effects

  • Electroporation for Primary Cells:

    • Specifically optimized for hard-to-transfect primary cells like CD4+ T lymphocytes

    • Successfully implemented for FKBP3 knockout in primary HIV-1 latency models

    • Requires careful optimization of electroporation parameters to balance transfection efficiency with cell viability

  • PROTAC (Proteolysis Targeting Chimera) Technology:

    • Emerging approach for targeted protein degradation

    • Could provide rapid, reversible, and dose-dependent depletion of FKBP3 protein

    • Particularly useful for acute intervention studies

For any FKBP3 modulation experiment, researchers should include appropriate controls:

  • Non-targeting sgRNAs or siRNAs

  • Empty vector controls

  • Rescue experiments with exogenous FKBP3 expression to confirm specificity

  • Monitoring of cell activation markers (CD25, CD69) and apoptosis to rule out non-specific effects

What cell models are most appropriate for studying FKBP3 functions in different contexts?

Selecting appropriate cell models is crucial for investigating FKBP3 functions across different research contexts:

For HIV-1 Latency Research:

  • Established Cell Line Models:

    • C11 cells: Contain HIV-1 proviral DNA with GFP reporter

    • J-Lat 10.6 cells: Widely used model with integrated but transcriptionally silent HIV-1

    • ACH2 cells: T-cell line with single copy of integrated HIV-1

    • Advantages: Stable, reproducible, easy to manipulate genetically

    • Limitations: May not fully recapitulate the complexity of latency in vivo

  • Primary Cell Models:

    • Primary CD4+ T lymphocytes: Isolated from multiple donors

    • Protocol highlights:

      • Infection with HIV-1 expressing NanoLuc luciferase

      • Gradual decrease in IL-2 levels to establish latency

      • Typically requires 12-14 days to develop latency

    • Advantages: Physiologically relevant, reflects donor variability

    • Limitations: More technically challenging, shorter experimental window

For Cancer Research (DLBCL):

  • DLBCL Cell Lines:

    • DB cells: Commonly used for FKBP3 studies in lymphoma

    • Other DLBCL lines (OCI-Ly1, OCI-Ly3, etc.) for validation

    • Advantages: Amenable to high-throughput screening and mechanistic studies

  • Xenograft Mouse Models:

    • DLBCL cells implanted into immunodeficient mice

    • Used to confirm in vitro findings on FKBP3's effect on tumor growth

    • Advantages: Provides in vivo validation of FKBP3's role in cancer progression

For Basic Molecular Function Studies:

  • HEK293T cells:

    • Useful for overexpression studies and protein-protein interaction analyses

    • Highly transfectable, suitable for biochemical characterization

  • Knockout Cell Lines:

    • CRISPR-generated FKBP3 knockout in relevant cell types

    • Creation of stable knockout lines enables comprehensive phenotyping

Technical Considerations for All Models:

  • Expression Verification:

    • Always confirm endogenous FKBP3 expression levels before selecting a model

    • Western blotting and qRT-PCR should be used to quantify expression

  • Functional Validation:

    • Ensure the model system expresses key FKBP3 interacting partners (YY1, HDAC1/2, PARK7)

    • Confirm relevant pathways (e.g., Wnt/β-catenin) are intact and responsive

  • Model-Specific Controls:

    • For HIV-1 latency: Monitor T-cell activation markers (CD25, CD69)

    • For cancer studies: Assess proliferation and apoptosis baselines

    • For all knockout studies: Include appropriate non-targeting controls

The selection of appropriate models should be guided by the specific research question, with consideration given to physiological relevance, technical feasibility, and the balance between in vitro and in vivo approaches.

How might FKBP3 inhibition be developed as a strategy for HIV-1 latency reversal?

Developing FKBP3 inhibition as a strategy for HIV-1 latency reversal represents a promising research direction with several considerations:

  • Target Validation Evidence:

    • FKBP3 knockout consistently activates latent HIV-1 in multiple cell models

    • The effect has been validated in physiologically relevant primary CD4+ T cell models

    • FKBP3 knockout does not significantly affect cell proliferation or apoptosis, suggesting a favorable safety profile

  • Potential Therapeutic Approaches:

    a) Small Molecule Inhibitors:

    • Design compounds targeting the PPIase domain of FKBP3

    • Consider structure-based drug design using available crystal structures (PDB ID: 5D75)

    • Develop selective inhibitors that distinguish FKBP3 from other FKBP family members

    • Could be combined with existing latency-reversing agents (LRAs) for synergistic effects

    b) Protein-Protein Interaction Disruptors:

    • Target the interaction between FKBP3 and YY1 or HDAC1/2

    • Peptide-based or small molecule approaches could disrupt the scaffolding function

    • This would prevent recruitment of HDACs to the HIV-1 LTR

    c) Antisense Oligonucleotides or siRNA:

    • Reduce FKBP3 expression through targeted RNA degradation

    • Could be delivered using nanoparticles targeted to CD4+ T cells

    • Temporary knockdown might be sufficient to induce latency reversal

  • Combination Therapy Potential:

    • FKBP3 inhibition could be combined with:

      • HDAC inhibitors (acting downstream in the same pathway)

      • PKC agonists (activating via different mechanisms)

      • Other LRAs targeting distinct pathways

    • This multi-pronged approach might achieve more complete latency reversal

  • Development Challenges:

    • Ensuring specificity for FKBP3 over other FKBP family members

    • Achieving sufficient target engagement in reservoir T cells

    • Minimizing off-target effects, particularly on immune function

    • Determining optimal dosing to achieve latency reversal without toxicity

  • Biomarker Development:

    • Histone acetylation levels at HIV-1 LTR could serve as pharmacodynamic markers

    • Development of assays to measure FKBP3 activity in patient samples

    • Identification of patient subgroups most likely to respond to FKBP3 targeting

The development of FKBP3-targeting approaches could potentially contribute to HIV-1 cure strategies by providing a new mechanism to reverse viral latency with potentially favorable safety profiles .

What are the potential implications of targeting FKBP3 in cancer therapy?

Targeting FKBP3 in cancer therapy presents several promising implications based on emerging research:

  • Therapeutic Potential in DLBCL:

    • FKBP3 aggravates malignant phenotypes of diffuse large B-cell lymphoma

    • Knockdown of FKBP3 has demonstrated:

      • Reduced proliferation of DLBCL cells

      • Decreased cancer cell stemness characteristics

      • Inhibited tumor growth in xenograft mouse models

    • These findings position FKBP3 as a potential therapeutic target for DLBCL treatment

  • Mechanism-Based Intervention Strategies:

    a) Disruption of FKBP3-PARK7 Interaction:

    • FKBP3 stabilizes PARK7 by preventing its ubiquitination and degradation

    • Targeting this specific protein-protein interaction could destabilize PARK7

    • This would subsequently inhibit Wnt/β-catenin signaling activation

    b) Wnt/β-catenin Pathway Modulation:

    • FKBP3 inhibition represents an upstream approach to target this pathway

    • Could be effective in cancers where direct Wnt inhibitors have shown limitations

    • May offer advantages in specificity compared to broader pathway inhibitors

    c) Combination with FOXO3 Activators:

    • FOXO3 naturally suppresses FKBP3 transcription

    • Therapies that activate FOXO3 could synergize with direct FKBP3 inhibitors

    • This dual approach might provide more durable responses

  • Broader Oncological Applications:

    • Beyond DLBCL, FKBP3 may have roles in other cancers through:

      • Epigenetic regulation via HDAC interactions

      • Transcriptional control through interactions with YY1 and other factors

      • Potential involvement in DNA damage response pathways

  • Therapeutic Development Considerations:

    • Selectivity: Developing compounds that specifically target FKBP3 while sparing other FKBP family members

    • Delivery: Strategies for lymphoma-specific delivery to minimize off-target effects

    • Resistance mechanisms: Understanding potential compensatory pathways that might emerge following FKBP3 inhibition

    • Biomarkers: Identifying patients most likely to respond to FKBP3-targeted therapy based on expression profiles

  • Safety Profile Assessment:

    • Initial research indicates FKBP3 knockout slightly increases cell proliferation but does not affect apoptosis

    • Comprehensive toxicology studies would be needed to establish the safety profile of FKBP3 inhibition

    • The specific role of FKBP3 in normal tissues must be thoroughly characterized

These findings collectively suggest that targeting FKBP3 represents a novel approach for cancer therapy, particularly in DLBCL, with potential applications in other malignancies where similar molecular mechanisms are active .

What techniques are available for high-throughput screening of FKBP3 modulators?

Several sophisticated high-throughput screening (HTS) approaches can be employed to identify and characterize FKBP3 modulators:

  • Biochemical Activity-Based Screens:

    a) PPIase Activity Assays:

    • Format: Measure cis-trans isomerization of proline-containing peptides

    • Detection: Fluorescence-based readouts using specially designed substrates

    • Advantages: Directly measures enzymatic function of FKBP3

    • Throughput: Can be miniaturized to 384 or 1536-well formats

    b) Thermal Shift Assays (TSA):

    • Format: Measure compound-induced changes in protein thermal stability

    • Detection: Fluorescent dyes bind to denatured proteins

    • Advantages: Requires minimal protein amounts, identifies direct binders

    • Applications: Initial screening and validation of hit compounds

  • Cell-Based Functional Screens:

    a) Reporter Gene Assays:

    • Design: Cells with HIV-1 LTR driving GFP or luciferase expression

    • Readout: Increased reporter expression indicates FKBP3 inhibition

    • Advantages: Identifies compounds that functionally reverse latency

    • Example: Modified J-Lat or C11 cells for FKBP3-focused screening

    b) Wnt/β-catenin Pathway Reporters:

    • Design: TOPFlash/FOPFlash luciferase reporters in DLBCL cells

    • Readout: Decreased signaling indicates effective FKBP3 inhibition

    • Applications: Cancer-focused screening for FKBP3 modulators

  • Protein-Protein Interaction Screens:

    a) AlphaScreen/AlphaLISA Technology:

    • Format: Bead-based proximity assay measuring interactions between FKBP3 and partners (YY1, HDAC1/2, PARK7)

    • Advantages: Homogeneous assay, high sensitivity, low background

    • Applications: Identify compounds that disrupt specific protein interactions

    b) Split Luciferase Complementation:

    • Design: FKBP3 and interacting partners fused to luciferase fragments

    • Readout: Decreased luminescence indicates disrupted interaction

    • Applications: Cell-based screening for interaction disruptors

  • Advanced Screening Technologies:

    a) CRISPR-Based Functional Genomics:

    • Approach: Genome-wide screens to identify synthetic lethal interactions with FKBP3

    • Applications: Discover combination strategies for FKBP3 inhibitors

    • Example: Previously used to identify FKBP3 as an HIV latency factor

    b) DNA-Encoded Libraries (DELs):

    • Approach: Screen billions of compounds against purified FKBP3 protein

    • Advantages: Massive compound diversity, minimal protein requirements

    • Output: Structure-activity relationships from single experiment

  • Data Analysis and Hit Characterization:

    a) Machine Learning Approaches:

    • Application: Analyze structure-activity relationships from primary screens

    • Advantage: Can predict properties of novel compounds based on training data

    b) Orthogonal Validation Assays:

    • Secondary biochemical assays to confirm mechanism of action

    • Cellular thermal shift assays (CETSA) to verify target engagement

    • Dose-response studies across multiple cell types to assess selectivity

These HTS approaches provide a comprehensive toolkit for identifying FKBP3 modulators, from initial discovery through hit validation and optimization. The combination of biochemical, cell-based, and advanced genomic techniques enables efficient identification of compounds with potential therapeutic applications in HIV latency reversal or cancer treatment .

What are the emerging roles of FKBP3 beyond HIV-1 latency and cancer?

FKBP3 is emerging as a multifunctional protein with roles extending beyond HIV-1 latency and cancer. Recent research has begun to uncover additional functions:

  • Epigenetic Regulation:

    • FKBP3's interactions with HDAC1/2 suggest a broader role in epigenetic regulation

    • The protein may function as a scaffold for chromatin-modifying complexes

    • This role could influence gene expression programs in various cellular contexts

    • The ability to bind DNA directly adds another dimension to its epigenetic functions

  • Cell Cycle and DNA Damage Response:

    • FKBP3's interaction with Mdm2 suggests involvement in p53 regulation

    • This may connect FKBP3 to cell cycle checkpoint control and DNA damage responses

    • Further investigation may reveal roles in genomic stability maintenance

  • Immune System Modulation:

    • As a member of the immunophilin family, FKBP3 likely has unexplored functions in immune regulation

    • The increased expression of FKBP3 observed upon HIV-1 infection of primary CD4+ T cells suggests active involvement in immune responses

    • This function may extend to other viral infections or inflammatory conditions

  • Protein Folding and Proteostasis:

    • The PPIase activity of FKBP3 suggests roles in protein folding

    • This function may be particularly important under cellular stress conditions

    • FKBP3 might participate in maintaining proteostasis in specialized cell types

  • Developmental Processes:

    • The involvement in fundamental cellular processes suggests potential roles in development

    • Wnt/β-catenin pathway regulation by FKBP3 (observed in cancer contexts) may have parallel functions in normal developmental signaling

  • Neurodegenerative Disease Connections:

    • FKBP3's interaction with PARK7 (DJ-1), which is associated with Parkinson's disease, suggests potential roles in neurodegeneration

    • The protein's function in protein folding may be relevant to proteinopathies characteristic of neurodegenerative conditions

These emerging roles highlight FKBP3 as a versatile cellular regulator involved in multiple fundamental processes. Future research focusing on these areas may reveal new therapeutic applications for FKBP3 modulation beyond the currently established roles in HIV-1 latency and cancer .

How can systems biology approaches advance our understanding of FKBP3 functions?

Systems biology approaches offer powerful frameworks for comprehensively understanding FKBP3's diverse functions and regulatory networks:

  • Multi-omics Integration:

    • Transcriptomics: RNA-seq analysis following FKBP3 modulation can reveal:

      • Global gene expression changes

      • Specific pathway perturbations

      • Direct vs. indirect regulatory effects

    • Proteomics: Mass spectrometry-based approaches can identify:

      • Complete FKBP3 interactome under various conditions

      • Post-translational modifications of FKBP3

      • Changes in protein complex formation

    • Epigenomics: ChIP-seq and ATAC-seq can map:

      • Genome-wide FKBP3 binding sites

      • Associated chromatin modifications

      • Accessibility changes following FKBP3 modulation

    • Integration of these datasets provides a holistic view of FKBP3 function across molecular levels

  • Network Analysis Approaches:

    • Protein-Protein Interaction Networks:

      • Place FKBP3 within its broader interaction ecosystem

      • Identify hub proteins connected to FKBP3

      • Reveal functional modules associated with FKBP3

    • Gene Regulatory Networks:

      • Map transcriptional effects of FKBP3 to regulatory circuits

      • Identify master regulators upstream and downstream of FKBP3

      • Model feedback loops involving FKBP3

  • Computational Modeling:

    • Dynamic Modeling:

      • Create mathematical models of FKBP3-dependent processes

      • Simulate temporal dynamics of HIV-1 latency establishment

      • Predict effects of perturbations on system behavior

    • Structural Bioinformatics:

      • Model FKBP3 interactions with partners at atomic resolution

      • Predict effects of mutations or drug binding

      • Identify allosteric mechanisms of regulation

  • Single-Cell Approaches:

    • Single-Cell RNA-seq:

      • Capture heterogeneity in FKBP3 expression and function

      • Identify cell populations particularly dependent on FKBP3

      • Track trajectories following FKBP3 modulation

    • Spatial Transcriptomics/Proteomics:

      • Map FKBP3 expression and effects in tissue context

      • Understand microenvironmental influences on FKBP3 function

  • Genome-Scale Functional Screening:

    • CRISPR Screens:

      • Identify synthetic lethal interactions with FKBP3

      • Discover genes that modify FKBP3-dependent phenotypes

      • Map genetic dependencies in FKBP3-high vs. FKBP3-low contexts

    • Drug Combination Screens:

      • Systematically test compounds that synergize with FKBP3 modulation

      • Build predictive models of drug-target-pathway interactions

  • Translational Systems Biology:

    • Patient Data Integration:

      • Correlate FKBP3 expression with clinical outcomes

      • Identify biomarkers predictive of response to FKBP3-targeting strategies

      • Stratify patient populations for personalized approaches

    • Multi-scale Modeling:

      • Connect molecular mechanisms to cellular, tissue, and organism-level outcomes

      • Predict therapeutic windows and optimal dosing strategies

These systems approaches collectively enable understanding FKBP3 not as an isolated protein but as a component within complex cellular networks. This comprehensive view can reveal emergent properties, identify novel therapeutic opportunities, and predict potential side effects of FKBP3 modulation that might not be apparent from reductionist approaches alone.

What are the most significant unresolved questions about FKBP3 biology?

Despite significant advances in FKBP3 research, several fundamental questions remain unanswered, representing important areas for future investigation:

  • Physiological Role and Regulation:

    • What is the primary physiological function of FKBP3 in normal cells?

    • How is FKBP3 expression and activity regulated under different cellular conditions?

    • What signals or stress conditions modulate FKBP3 levels and localization?

    • Why does HIV-1 infection increase FKBP3 expression in primary CD4+ T cells?

  • Structural Biology Questions:

    • What conformational changes occur when FKBP3 interacts with different binding partners?

    • How does FKBP3 simultaneously coordinate interactions with multiple partners (YY1, HDAC1/2)?

    • What is the structural basis for FKBP3's higher affinity for rapamycin compared to FK506?

    • How does the DNA-binding capacity of FKBP3 structurally coordinate with its protein interactions?

  • Mechanistic Uncertainties:

    • Is the PPIase activity of FKBP3 required for its role in HIV-1 latency and cancer progression?

    • Does FKBP3 have additional enzymatic functions beyond prolyl isomerization?

    • How does FKBP3 specifically recognize its DNA binding sites?

    • What determines the specificity of FKBP3 for certain promoter regions like the HIV-1 LTR?

  • Therapeutic Target Development:

    • Can FKBP3 be selectively inhibited without affecting other FKBP family members?

    • What are the potential side effects of long-term FKBP3 inhibition?

    • Are there natural or synthetic ligands that modulate FKBP3 function?

    • How might resistance mechanisms develop against FKBP3-targeted therapies?

  • Broader Biological Context:

    • What is the evolutionary significance of FKBP3's diverse functions?

    • Are there undiscovered roles for FKBP3 in other viral infections beyond HIV-1?

    • Does FKBP3 contribute to other disease states through mechanisms similar to those in HIV-1 latency and DLBCL?

    • How does FKBP3 function differ across cell types and developmental stages?

  • Clinical Relevance:

    • Do FKBP3 expression levels correlate with HIV-1 reservoir size or stability in patients?

    • Can FKBP3 serve as a biomarker for specific cancer subtypes or treatment response?

    • Are there natural polymorphisms in the FKBP3 gene that affect disease susceptibility or progression?

    • How does FKBP3 interact with current therapeutic agents for HIV or cancer?

  • Systems-Level Understanding:

    • What are the comprehensive network effects of FKBP3 modulation across different cellular contexts?

    • How does FKBP3 function integrate with other epigenetic regulators to establish and maintain specific chromatin states?

    • Are there feedback loops that regulate FKBP3 function in response to its own activity?

Addressing these unresolved questions will require interdisciplinary approaches combining structural biology, biochemistry, cell biology, systems biology, and clinical research. Resolving these uncertainties could significantly advance our understanding of FKBP3 biology and accelerate its development as a therapeutic target for HIV-1 latency reversal and cancer treatment .

Product Science Overview

Structure and Function

FKBP3 is characterized by its peptidyl-prolyl cis-trans isomerase (PPIase) activity, which facilitates protein folding by catalyzing the isomerization of proline residues in polypeptides . This activity is crucial for the proper folding and function of many proteins within the cell.

In addition to its PPIase activity, FKBP3 has been found to play a role in various cellular processes, including:

  • Protein Folding: FKBP3 acts as a chaperone, assisting in the proper folding of newly synthesized proteins .
  • DNA Repair: FKBP3 has been shown to bind non-specifically to DNA and is involved in DNA repair mechanisms .
  • Signal Transduction: By forming complexes with other proteins, FKBP3 can influence various signaling pathways within the cell .
Role in Immunosuppression

One of the most notable functions of FKBP3 is its role in immunosuppression. When bound to FK506, FKBP3 forms a complex that inhibits the phosphatase activity of calcineurin . This inhibition prevents the activation of the nuclear factor of activated T-cells (NF-AT), thereby blocking T-cell activation and proliferation. This mechanism is particularly important in preventing organ rejection in transplant patients and in treating autoimmune disorders .

Research and Therapeutic Applications

FKBP3 and other members of the FKBP family have become subjects of considerable interest in various fields of research due to their involvement in numerous cellular and molecular pathways . Some key areas of research and therapeutic applications include:

  • Cancer Research: FKBP3 has been implicated in the regulation of cell growth and apoptosis, making it a potential target for cancer therapy .
  • Neurodegenerative Diseases: The role of FKBP3 in protein folding and stress response pathways suggests its potential involvement in neurodegenerative diseases such as Alzheimer’s and Parkinson’s .
  • Drug Development: Understanding the binding mechanisms of FK506 and rapamycin to FKBP3 can aid in the development of new immunosuppressive drugs and other therapeutic agents .

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 2024 Thebiotek. All Rights Reserved.