RANK Mouse refers to genetically modified murine models (e.g., knockouts, transgenics) used to study the RANK/RANKL pathway. Key genetic variants include:
RANK −/− mice lack functional osteoclasts, leading to osteopetrosis (excessive bone density) and splenic B cell deficiency .
RANKL inhibition (via RANK-Fc) in oim/oim mice improved femoral cortical thickness (+18%) and stiffness (+25%) compared to saline-treated controls .
RANK deficiency abolishes peripheral lymph node formation but preserves mucosal-associated lymphoid tissue .
Dendritic cell (DC) differentiation and macrophage function remain intact in RANK −/− mice, indicating RANK’s specificity for osteoclast and lymph node pathways .
Cytokine-induced β cell death in mice and human islets requires RANK-TRAF6 interaction and NF-κB activation.
Denosumab (RANKL antibody) reduced β cell apoptosis by 40–60% in vitro and enhanced proliferation in transplanted human islets .
MMTV-RANK transgenic mice showed RANKL-dependent mammary epithelial cell (MEC) proliferation and suppressed differentiation:
The RACE assay estimates drug exposure (eAUC<sub>20–120</sub>) in mice using a standardized protocol (n=4 mice, single dose) .
Simplifies pharmacokinetic profiling for RANK pathway inhibitors like Denosumab .
Collaborative Cross (CC) and Diversity Outbred (DO) mice capture 90% of Mus musculus genetic diversity, enabling nuanced RANK pathway analysis .
Tumor necrosis factor receptor superfamily member 11A, Osteoclast differentiation factor receptor, ODFR, Receptor activator of NF-KB, activator of NFKB, FEO, OFE, OSTS, PDB2, RANK, RANKLOH18CR1, CD265, CD265 antigen, OPTB7, TRANCER, LOH18CR1, receptor activator of nuclear factor-kappa B.
Sf9, Baculovirus cells.
ADLVTPPCTQ ERHYEHLGRC CSRCEPGKYL SSKCTPTSDS VCLPCGPDEY LDTWNEEDKC LLHKVCDAGK ALVAVDPGNH TAPRRCACTA GYHWNSDCEC CRRNTECAPG FGAQHPLQLN KDTVCTPCLL GFFSDVFSST DKCKPWTNCT LLGKLEAHQG TTESDVVCSS SMTLRRPPKE AQAYLPSLEP KSCDKTHTCP PCPAPELLGG PSVFLFPPKP KDTLMISRTP EVTCVVVDVS HEDPEVKFNW YVDGVEVHNA KTKPREEQYN STYRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPSRDE LTKNQVSLTC LVKGFYPSDI VEWESNGQP ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT QKSLSLSPGK HHHHHH
RANK Ligand (Receptor Activator of Nuclear Factor kappa-B Ligand) is a cell-bound marker related to the tumor necrosis factor (TNF) family of proteins. It plays a critical role in bone metabolism and osteoclast differentiation. In mouse models, RANK Ligand expression by T cells promotes dendritic cell maturation, making it an important molecule for studying both skeletal and immune system interactions . Mouse models using recombinant RANK Ligand have been instrumental in understanding bone diseases, immune disorders, and cancer metastasis processes.
Reproducible RANK mouse research requires careful attention to experimental design principles. The Jackson Laboratory recommends several key strategies to enhance reproducibility:
Choose mouse strains wisely: Different strains have unique characteristics that affect RANK/RANKL expression and function
Calculate appropriate sample sizes based on power analyses to detect biologically relevant differences
Implement proper randomization and blinding procedures
Control for environmental variables that affect RANK/RANKL signaling
Standardize protocols for tissue collection, processing, and analysis
Additionally, follow the National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs) principles, which provide a framework for ethical animal use and promote robust experimental design. Remember that even genetically identical mice within a particular strain can show phenotypic variability, are sensitive to environmental factors, and change developmentally over time .
Strain selection significantly influences experimental outcomes in RANK/RANKL research. Consider these factors:
C57BL/6J mice are widely used due to their well-characterized genome and immunological responses
Different strains develop age-related bone phenotypes at varying rates, affecting RANK/RANKL signaling interpretation
Genetic background effects on bone phenotype can mask or enhance RANK/RANKL-related outcomes
For transgenic and knockout studies, backcrossing to a consistent genetic background for at least 10 generations minimizes genetic variability
As The Jackson Laboratory notes, "Mouse strains are as variable as dog breeds, if not more so," making strain selection a critical experimental variable .
Proper handling of recombinant mouse RANK Ligand is essential for experimental success:
Centrifuge the vial before opening
Reconstitute by gently pipetting the recommended solution down the sides of the vial
DO NOT VORTEX the solution
Allow several minutes for complete reconstitution
For prolonged storage, dilute to working aliquots in a 0.1% BSA solution
Failure to follow these guidelines may result in loss of protein activity and experimental variability.
Multiple complementary techniques provide comprehensive assessment of RANK/RANKL expression:
Quantitative PCR (qPCR): Provides sensitive detection of mRNA expression with proper reference gene selection
Western Blotting: Confirms protein expression using validated antibodies specific to mouse RANK/RANKL
Immunohistochemistry: Provides spatial information on expression patterns with appropriate controls
Flow Cytometry: Enables quantification at the single-cell level, particularly useful for immune cell populations
ELISA: Quantifies soluble RANKL in serum or culture media
For optimal results, combine at least two of these approaches to verify expression at both mRNA and protein levels.
The following protocol optimizes osteoclast precursor isolation and culture:
Isolation from bone marrow:
Harvest femurs and tibias from 6-12 week old mice
Flush bone marrow with cold PBS using a 25G needle
Culture in α-MEM with 10% FBS and M-CSF (25-50 ng/mL) for 3 days
Harvest non-adherent cells, which contain enriched osteoclast precursors
Culture conditions for osteoclastogenesis:
Base medium: α-MEM with 10% heat-inactivated FBS
Essential cytokines: M-CSF (25-50 ng/mL) and RANKL (50-100 ng/mL)
Plating density: 5×10⁴ cells/cm² for optimal fusion
Culture duration: 5-7 days for mature osteoclasts
Verification: TRAP staining and counting multinucleated (≥3 nuclei) TRAP+ cells
This approach typically yields approximately 2-3×10⁷ precursor cells per mouse.
The Mouse Positioning and Surveillance System (MoPSS) offers an effective approach for preference testing in laboratory mice. This system consists of:
Two interconnected cages with a connecting plexiglass tube
RFID antennas attached to the tube to track individual mouse movement
Plastic barriers within the tube to slow mouse movement and ensure RFID detection
For optimal experimental design:
House mice in groups of 4 animals to maintain normal social behavior
Use identical environmental conditions (light, temperature, humidity) for testing
Ensure equal light distribution using LED lighting aimed at the ceiling
Monitor light intensity with a lux meter to verify consistency
Equip both cages with standard bedding, shelter, nesting materials, and food/water
Conduct tests over 46 hours to capture both active and inactive phases
Analysis involves calculating stay times for each mouse in each cage, expressed as a percentage of the total testing period. This methodology allows assessment of individual and group preferences for different enrichment items.
The "worth value" calculation is an advanced statistical approach for ranking multiple enrichment items based on mouse preferences. This methodology:
Combines data from multiple binary choice tests where mice choose between different enrichment items
Calculates a relative preference score ("worth value") for each item
Normalizes these values across all tested items
This approach is particularly valuable when categorizing enrichment items by their intended purpose:
Structural enrichment (physical cage organization)
Foraging enrichment (items stimulating natural food-seeking behaviors)
The worth value method offers a more robust assessment than simple binary comparisons and accounts for individual variability in preference strength.
Distinguishing between these RANKL forms is critical as they may have different biological activities:
Methodological approaches:
Differential centrifugation:
Sequential centrifugation separates membrane fractions containing membrane-bound RANKL
Ultracentrifugation (>100,000g) isolates membrane vesicles
Western blotting of fractions can confirm distribution
Flow cytometry:
Surface staining without permeabilization detects membrane-bound RANKL
Compare with permeabilized samples to assess total RANKL
Use fluorescence-minus-one controls for accurate gating
ELISA with sample preparation modifications:
Pretreatment of samples with detergents releases membrane-bound RANKL
Compare treated vs. untreated samples to quantify the membrane-bound fraction
Transgenic approaches:
Mouse models expressing only membrane-bound RANKL (deletion of cleavage site)
Conditional knockout models for tissue-specific expression analysis
Statistical analysis should be tailored to the specific experimental design and data characteristics:
For comparing genotypes/treatments:
Two-group comparisons: t-test (parametric) or Mann-Whitney (non-parametric)
Multiple group comparisons: ANOVA with appropriate post-hoc tests
Tukey's test for all pairwise comparisons
Dunnett's test when comparing to a control group
Repeated measures designs: RM-ANOVA or mixed-effects models
Sample size considerations:
For gene expression studies: n=8-10 per group
For histomorphometry: n=6-8 per group
For serum biomarkers: n=10-12 per group
Reporting standards:
Include exact p-values rather than thresholds
Report effect sizes and confidence intervals
Present individual data points in addition to means/medians
Clearly state the statistical tests used and software version employed
Normalization strategies should be tailored to the detection method and sample types:
For qPCR data:
Use multiple reference genes validated for stability across experimental conditions
Recommended reference gene combinations by tissue type:
Bone: Actb, Hprt1, and Tbp
Immune cells: Rpl13a, Ppia, and Gapdh
Apply geometric averaging of multiple reference genes
For protein quantification:
Western blotting: Normalize to total protein (Ponceau, REVERT) rather than single housekeeping proteins
ELISA: Express as concentration per unit protein or per tissue weight
Flow cytometry: Use median fluorescence intensity ratio to isotype control
For histological quantification:
Express as positive cells per defined area or tissue volume
Use stereological approaches for unbiased quantification
Contradictory findings require systematic evaluation of multiple factors:
Strain-dependent effects:
C57BL/6 substrains may respond differently (J vs. N backgrounds)
BALB/c mice have different baseline bone turnover than C57BL/6
DBA/2 mice show heightened sensitivity to mechanical loading
Age and sex considerations:
Young mice (4-8 weeks): Rapid growth, high bone turnover
Adult mice (12-16 weeks): Stable bone mass, moderate turnover
Aged mice (>12 months): Age-related bone loss, altered RANKL sensitivity
Males vs. females: Estrogen modulates RANKL signaling
Methodological divergences:
Dose and duration of RANKL administration
Route of administration (local vs. systemic)
Protein source (commercial vendor, purification method)
Timing of measurements relative to intervention
Reconciliation strategies:
Direct replication studies with standardized protocols
Meta-analysis of multiple studies with moderator variables
Investigation of contextual factors (diet, housing, microbiome)
Biological variability in RANK/RANKL systems can be managed through several approaches:
Sources of variability:
Genetic: Even inbred strains show some variation
Environmental: Housing conditions, handling stress, microbiome
Developmental: Age, sex, reproductive status
Technical: Sample collection, processing, analysis methods
Experimental design strategies:
Increase sample size based on power calculations from preliminary data
Use paired designs where possible (e.g., contralateral limbs)
Block randomization by litter, cage, or initial body weight
Include baseline measurements as covariates in analysis
Standardization protocols:
Consistent time of day for sample collection (circadian effects)
Uniform fasting periods before blood collection
Standardized anesthesia protocols
Consistent tissue harvesting and processing times
When using systems like the Mouse Positioning and Surveillance System (MoPSS), several factors can lead to inconsistent results:
Technical considerations:
RFID antenna sensitivity and placement
Detector calibration and maintenance
Data logging system integrity
Biological factors:
Individual mouse personality/temperament differences
Social hierarchy effects within groups
Previous enrichment exposure history
Age and sex of test subjects
Environmental variables:
Light intensity differences between cages
Airflow direction and intensity
External noises or vibrations
Temperature gradients
Analysis considerations:
Accounting for both active and inactive phases (full 46-hour analysis)
Distinguishing individual vs. group preferences
Proper statistical handling of missing data points
Distinguishing direct from indirect effects requires specialized experimental approaches:
Cell-specific genetic models:
Cre-loxP systems with cell-specific promoters:
Lysozyme M-Cre for myeloid lineage
Cathepsin K-Cre for mature osteoclasts
Col1a1-Cre for osteoblasts
CD4-Cre for T lymphocytes
Tamoxifen-inducible systems to control timing of gene deletion
Ex vivo approaches:
Isolated cell cultures with defined conditions
Co-culture systems with different cell types separated by permeable membranes
Conditioned medium experiments to identify secreted factors
Cell-specific inhibitors or neutralizing antibodies
Bone marrow chimeras:
Irradiation and reconstitution with donor cells
Mixed chimeras with wild-type and mutant cells
Cell-specific tracking using congenic markers (CD45.1/CD45.2)
Molecular approaches:
Phosphorylation state analysis for direct signaling targets
Temporal gene expression analysis to establish sequence of events
ChIP-seq for identifying direct transcriptional targets
RANK is a type I transmembrane protein that is expressed on the surface of various cell types, including osteoclast precursors, dendritic cells, and mammary gland epithelial cells. The extracellular domain of RANK binds to its ligand, RANKL (Receptor Activator of Nuclear Factor Kappa-Β Ligand), which is also known as TRANCE, TNFSF11, or OPGL .
The primary function of RANK is to mediate the effects of RANKL. When RANKL binds to RANK, it triggers a signaling cascade that leads to the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and other downstream pathways. This signaling is essential for:
Recombinant mouse RANK is produced using various expression systems, such as E. coli. The recombinant protein is typically purified to high levels of purity and is used in various research applications, including:
Recombinant mouse RANK is used extensively in research to understand its role in bone metabolism and immune regulation. It is also used to screen potential inhibitors that could be developed into drugs for treating bone-related diseases and immune disorders. The ability to produce recombinant RANK in a controlled environment allows researchers to conduct detailed studies on its structure, function, and interactions with other proteins .