FK506-binding protein like (FKBPL), encoded by the FKBPL gene, is a 349-amino-acid immunophilin-like protein with structural homology to FK506-binding proteins (FKBPs). It was initially identified as DIR1 (DNA damage-inducible transcript 1) and later renamed FKBPL due to its functional similarities to FKBP family members . Key features include:
Aliases: DIR1, NG7, Wisp39 (WAF-1/CIP1 stabilizing protein 39) .
Structural Domains: Contains a tetratricopeptide repeat (TPR) domain critical for protein-protein interactions .
Interactions: Binds Hsp90, glucocorticoid receptors, and dynamitin, influencing signaling pathways and protein stability .
Primary Functions:
Stress Response: Regulates cellular adaptation to stressors like radiation .
Cell Cycle Control: Stabilizes p21 (a cyclin-dependent kinase inhibitor), delaying G1/S phase transition .
Angiogenesis: Acts as a secreted anti-angiogenic factor targeting CD44 .
Apoptosis: Modulates pro- and anti-apoptotic proteins, such as HSP27 and phospho-p53 .
FKBPL lacks a classical signal peptide but is secreted via non-conventional pathways. Its TPR domain facilitates interactions with:
Intracellular:
Extracellular:
FKBPL suppresses angiogenesis via CD44-mediated signaling, inhibiting endothelial sprouting and tumor vascularization . Key findings:
Peptide | Target | Phase | Key Findings |
---|---|---|---|
ALM201 | CD44 | Phase 1a completed | Safe in humans; orphan designation for ovarian cancer |
AD-01 | CD44-independent | Preclinical | Reduces cytokines (TNF, IL-6) in LPS models |
FKBPL regulates inflammation through NF-κB modulation:
Condition | FKBPL Effect | Mechanism |
---|---|---|
Sepsis | Reduces TNF/IL-6 production | Inhibits p65 phosphorylation |
Endothelial stress | Enhances VE-cadherin junctions | Maintains barrier integrity |
A study of 222 lung adenocarcinoma patients demonstrated:
METPPVNTIG EKDTSQPQQE WEKNLRENLD SVIQIRQQPR DPPTETLELE VSPDPASQIL EHTQGAEKLV AELEGDSHKS HGSTSQMPEA LQASDLWYCP DGSFVKKIVI RGHGLDKPKL GSCCRVLALG FPFGSGPPEG WTELTMGVGP WREETWGELI EKCLESMCQG EEAELQLPGH SGPPVRLTLA SFTQGRDSWE LETSEKEALA REERARGTEL FRAGNPEGAA RCYGRALRLL LTLPPPGPPE RTVLHANLAA CQLLLGQPQL AAQSCDRVLE REPGHLKALY RRGVAQAALG NLEKATADLK KVLAIDPKNR AAQEELGKVV IQGKNQDAGL AQGLRKMFGL EHHHHHH.
FKBPL (FK506-binding protein like) is a member of the immunophilin protein superfamily with potent anti-tumor activity primarily through inhibition of angiogenesis and cancer stemness. Also known as DIR1, NG7, and WAF-1/CIP1 stabilizing protein 39 (WISp39), FKBPL contains 349 amino acids in humans and features a unique N-terminal region that confers its biological activity. Unlike some immunophilins, FKBPL contains a non-functional PPIase domain in this N-terminal region, which is independent of its TPR domains but critical for its biological effects .
The full amino acid sequence of human FKBPL protein (1-349) is:
METPPVNTIGEKDTSQPQQEWEKNLRENLDSVIQI RQQPRDPPTETELELEVSPDPASQILEHTQGAEKLV AELEGDSHKSHGSTSQMPEALQASDLWYCPDGSFV KKIVIRGHGLDKPKLGSCCRVLALGFPFGSGPPEG WTELTMGVGPWEREETWGELIEKCLESMCQGEEA ELQLPGHSGPPVRLTLASFTQGRDSWELETSEKEA LAREERARGTELFRAGNPEGAARCYGRALRLLLTL PPPGPPERTV LHANLAACQLLLGQPQLAAQS CDRVLEREPGHLKALYRRGVAQAALGN LEKATAD LKKVLAIDPKNRAAQEELGKVVIQGKNQDAGLAQG LRKMF
Methodology note: When working with recombinant FKBPL, researchers often use the His-tagged version expressed in Escherichia coli with >90% purity, which is suitable for SDS-PAGE, mass spectrometry, and functional studies .
FKBPL has several important cellular functions:
Regulation of vascular integrity through modulation of endothelial cell barrier function
Negative regulation of NF-κB activation and inflammatory signaling
Anti-angiogenic activity through CD44-dependent mechanisms
Anti-cancer stem cell (CSC) activity by regulating expression of pluripotency transcription factors
Regulation of p21 protein stability through binding to Hsp90 and p21
Involvement in cellular response to X-ray radiation
When studying these functions, researchers should implement appropriate controls and utilize both gain-of-function and loss-of-function approaches to validate findings .
For effective FKBPL knockdown, siRNA transfection has been successfully used in human microvascular endothelial cells (HMEC-1), achieving approximately 80% reduction in both mRNA and protein expression. This technique is particularly useful for investigating FKBPL's role in endothelial barrier function and inflammatory responses.
Methodological considerations:
Validate knockdown efficiency by measuring both mRNA (qPCR) and protein (Western blot) levels
Include appropriate non-targeted siRNA controls
Consider the impact of inflammatory stimuli (e.g., LPS) on knockdown efficiency
For phenotypic assays after knockdown, an in vitro Evan's blue permeability assay can effectively measure changes in endothelial barrier function .
To investigate FKBPL's role in NF-κB signaling:
Isolate bone marrow-derived macrophages (BMDMs) from wild-type and Fkbpl+/- mice
Stimulate cells with LPS (100 ng/ml) for different time points (e.g., 45, 60, 90 minutes)
Assess phosphorylation of p65(RelA) via Western blotting
For peptide studies, include AD-01 (1 nM) in treatment groups
To determine CD44 dependency, use CD44 knockout mouse models
Include controls for other inflammatory signaling pathways (e.g., JNK phosphorylation)
This approach allows for rigorous assessment of FKBPL's specific effects on NF-κB signaling while controlling for potential confounding effects on other inflammatory pathways .
Several in vivo models have proven valuable for FKBPL research:
Fkbpl+/- haploinsufficient mice:
Useful for studying developmental angiogenesis
Show vascular irregularities and enhanced angiogenesis
Display increased erythrocyte leakage into surrounding tissues
Demonstrate reduced survival in LPS-induced inflammatory challenge
LPS-induced inflammatory models:
Effective for testing FKBPL peptides (AD-01, ALM201) in vivo
Allow assessment of survival outcomes and inflammatory cytokine production
Permit collection of peritoneal lavage for cytokine analysis
CD44 knockout mice (CD44-/-):
Valuable for determining CD44-dependency of FKBPL mechanisms
Can be used to differentiate between angiogenic and inflammatory pathways
When designing in vivo experiments, researchers should carefully consider dosing regimens, timing of interventions, and appropriate controls to account for genetic background effects .
FKBPL functions as a critical regulator of vascular integrity, particularly during inflammatory responses. When FKBPL is knocked down in human microvascular endothelial cells (HMEC-1), there is a significant increase in barrier permeability (p = 0.0347), which is further exacerbated by LPS stimulation. This indicates that FKBPL plays a protective role in maintaining endothelial barrier function.
At the molecular level, FKBPL regulates vascular integrity through:
Modulation of VE-cadherin endothelial tight junctions
Inhibition of NF-κB signaling in endothelial cells
Regulation of inflammatory cytokine production
Importantly, treatment with the FKBPL-based peptide AD-01 increases VE-cadherin endothelial tight junctions following LPS stimulation, offering a potential therapeutic approach for stabilizing vascular barriers during inflammatory conditions .
FKBPL shares functional similarities with other immunophilins in regulating NF-κB signaling, but through distinct mechanisms:
FKBP51 complexes with cytoplasmic p65 in unstimulated cells
Upon stimulation, FKBP51 is exchanged for FKBP52
FKBP52 is then recruited to the promoter region of NF-κB target genes
Important distinctions:
The TPR domains of FKBP51 and FKBP52 are not required for NF-κB regulation
FKBP52's PPIase activity is required for its NF-κB stimulatory function
FKBP51's PPIase activity is not required for its inhibitory action
FKBPL peptides (AD-01, ALM201) based on the N-terminal region (containing the non-functional PPIase domain) modulate NF-κB similarly to full-length FKBPL
These findings suggest that despite structural differences, FKBPL regulates NF-κB through its N-terminal domain, similar to but distinct from other immunophilins. Further research is needed to fully understand the interaction between FKBPL, FKBP51, and FKBP52 in NF-κB regulation .
Genetic analysis has revealed significant associations between FKBPL variants and human inflammatory and autoimmune conditions:
Methodology for genetic association studies:
Utilize global biobank engines comprising multiple geographical biobanks
Assess variants in fkbpl, fkbp51, and fkbp52 associated with phenotypes of interest
Apply genome-wide association study (GWAS) significance threshold of p≤ 5×10^-8
Analyze tissue-specific gene expression levels using databases like GTEx
Investigate disease-specific expression using microarray data from resources like NCBI GEO
Findings from such analyses have revealed associations between FKBPL genetic variants and:
Psoriasis
Rheumatoid arthritis
High lymphocyte count
Potentially other inflammatory disorders
These genetic associations support FKBPL's physiological role in regulating inflammatory responses and suggest genetic testing for FKBPL variants could help identify individuals at risk for inflammatory conditions .
FKBPL-based peptides have shown remarkable potential as therapeutic agents:
AD-01: A 24-residue peptide (amino acids 34-58 of FKBPL)
Pre-clinical peptide with potent anti-tumor activity
Inhibits p65(RelA) phosphorylation following LPS stimulation
Reduces NF-κB target gene expression and pro-inflammatory cytokine production
Functions independently of CD44 in regulating NF-κB signaling
ALM201: A more stable 23-residue peptide
Clinical drug candidate with equipotent activity to AD-01
Completed Phase 1a dose-escalation clinical trial in patients with ovarian cancer and other solid tumors
Demonstrated favorable pharmacokinetic and safety profile with no major adverse events
Received orphan drug status from FDA for ovarian cancer
In LPS survival models, treatment resulted in 100% survival rate at experimental endpoint
Abrogated production of pro-inflammatory cytokines (TNF and IL-6) in peritoneal lavage washings
These peptides represent a significant advancement in developing FKBPL-based therapeutics that could address both vascular instability and excessive inflammation in conditions such as sepsis, acute respiratory distress syndrome (ARDS), and potentially severe COVID-19 .
Recent research has identified FKBPL as a potential biomarker and therapeutic target in metabolic and cardiovascular conditions:
A cross-sectional study of 353 adults (234 with Type 2 diabetes and 119 non-diabetic subjects) found:
Higher plasma FKBPL levels in Type 2 diabetes patients (adjusted mean: 2.03 ng/ml ± 0.90 SD) compared to non-diabetic subjects (adjusted mean: 1.79 ng/ml ± 0.89 SD)
Association between FKBPL levels and cardiovascular disease risk
These findings suggest:
FKBPL may play a role in the pathophysiology linking diabetes and cardiovascular complications
Disturbed angiogenesis and endothelial dysfunction, both regulated by FKBPL, are implicated in both conditions
FKBPL could potentially serve as a biomarker for cardiovascular risk in diabetic patients
Researchers investigating FKBPL in metabolic disorders should employ appropriate case-control designs with matching for age, BMI, and gender to minimize confounding factors .
FKBPL-based therapeutics show promise for treating a range of inflammatory conditions:
Acute inflammatory conditions:
Sepsis
Acute respiratory distress syndrome (ARDS)
Severe COVID-19
Other conditions characterized by cytokine storm
Chronic inflammatory diseases:
Psoriasis
Rheumatoid arthritis
Potentially other autoimmune conditions
The dual activity of FKBPL-based peptides in:
Stabilizing vascular barriers (reducing leakage)
Inhibiting pro-inflammatory cytokine production
Makes them particularly valuable candidates for conditions where both vascular dysfunction and excessive inflammation contribute to pathology. The favorable safety profile demonstrated in clinical trials further enhances their therapeutic potential .
When faced with contradictory findings in FKBPL research, scientists should consider:
Context-dependent effects:
FKBPL may have different functions in different cell types (e.g., endothelial cells vs. macrophages)
Inflammatory state may affect FKBPL's role (baseline vs. stimulated conditions)
Interaction with different binding partners in different tissues
Methodological differences:
In vitro vs. in vivo models
Acute vs. chronic interventions
Genetic models vs. pharmacological approaches
Differences in experimental conditions (e.g., LPS concentration, timing)
Experimental approaches to resolve contradictions:
Use multiple complementary techniques to study the same phenomenon
Include appropriate positive and negative controls
Validate findings across different cell types and experimental models
Consider dose-response relationships rather than single-dose experiments
Perform time-course studies to capture dynamic changes
By systematically addressing these factors, researchers can better understand the complex and context-dependent functions of FKBPL .
For accurate measurement of FKBPL in human samples:
Plasma/Serum quantification:
ELISA has been successfully used to measure FKBPL levels in plasma from diabetic and non-diabetic subjects
Important to standardize collection, processing, and storage of samples
Tissue expression:
Immunohistochemistry can assess FKBPL expression in tissue samples
qPCR for mRNA quantification
Western blotting for protein expression
Genetic analysis:
Genotyping of FKBPL variants using next-generation sequencing
Analysis of expression quantitative trait loci (eQTLs) using databases like GTEx
Methodological considerations:
Include appropriate controls for each assay
Account for potential confounding factors (age, gender, BMI, medications)
Validate findings using multiple methodological approaches
Consider tissue-specific expression patterns when interpreting results
Researchers should select methods based on their specific research questions and available sample types .
FK506 Binding Proteins (FKBPs) are a family of proteins that have garnered significant interest due to their role in various cellular processes and their interaction with immunosuppressive drugs such as FK506 (tacrolimus) and rapamycin . These proteins are known for their peptidyl-prolyl cis-trans isomerase (PPIase) activity, which aids in protein folding and function .
FKBPs are characterized by their ability to bind to FK506 and rapamycin, forming complexes that inhibit the phosphatase activity of calcineurin . This inhibition is crucial for the immunosuppressive effects of these drugs, particularly in preventing organ rejection post-transplantation . The FKBP family includes several members, each with distinct functions and tissue distributions .
One of the most studied members of this family is FKBP12. It binds to FK506 and rapamycin, inhibiting calcineurin and thus blocking T-cell activation . FKBP12 is also involved in the regulation of calcium channels, such as the ryanodine receptor, which plays a critical role in calcium signaling pathways .
The unique properties of FKBPs have made them valuable tools in biological research. For instance, FKBP12 can be used in chemically induced dimerization applications to manipulate protein localization and signaling pathways . Additionally, the immunosuppressive properties of FKBP-binding drugs have been harnessed in clinical settings to prevent organ rejection and treat autoimmune diseases .