Recombinant Mouse Tumor necrosis factor ligand superfamily member 11 (Tnfsf11), partial (Active)

Shipped with Ice Packs
In Stock

Description

Functional Activity and Bioassay Data

TNFSF11’s primary role is osteoclast differentiation and activation via RANK signaling. Key bioactivity findings include:

Osteoclastogenesis

  • RAW 264.7 Cell Line: Recombinant TNFSF11 induces osteoclast differentiation at ED₅₀ values of 5–15 ng/mL (mammalian systems) or 0.5–2 ng/mL (E. coli systems), depending on the need for cross-linking antibodies .

  • Mechanism: Activates NF-κB, Ca²⁺ oscillations, and CREB1 pathways, leading to osteoclast-specific gene transcription .

Immune Regulation

  • T Cell and Dendritic Cell Function: Enhances T cell proliferation and dendritic cell survival, critical for adaptive immune responses .

  • Lymph Node Organogenesis: Essential for lymphoid tissue development .

Research Applications

TNFSF11 is utilized in diverse experimental contexts:

ApplicationDetailsSources
Osteoclast Differentiation AssaysInduces RAW 264.7 or bone marrow-derived macrophage differentiation into mature osteoclasts
ELISA StandardsServes as a calibration control for quantifying TNFSF11 levels in biological samples
Western Blot ControlsValidates antibody specificity for TNFSF11 detection
Transgenic StudiesBAC transgenes containing the Tnfsf11 locus restore osteoclastogenesis and immune cell development in knockout mice

Regulatory Factors and Physiological Relevance

TNFSF11 expression is tightly regulated by:

FactorEffectSource
1,25-Dihydroxyvitamin D₃Upregulates TNFSF11 transcription in osteoblasts and immune cells
Parathyroid Hormone (PTH)Induces TNFSF11 expression in bone and mammary tissues
Lipopolysaccharide (LPS)Activates TNFSF11 via TLR4 signaling in immune cells and osteoblasts

These regulatory pathways are critical for understanding TNFSF11’s role in calcium metabolism, immune surveillance, and disease states like osteoporosis .

Therapeutic and Pathological Implications

Overexpression of TNFSF11 is linked to:

  • Bone Diseases: Rheumatoid arthritis, psoriatic arthritis, and medication-related osteonecrosis .

  • Immune Dysregulation: Impaired T/B cell development in Tnfsf11 knockout mice .

Conversely, TNFSF11 inhibition (e.g., via OPG or denosumab) is therapeutic in osteolytic disorders .

Key Research Findings

  1. Transgenic Rescue: A Tnfsf11 BAC transgene restored osteoclastogenesis and immune cell development in Tnfsf11−/− mice, confirming the locus’s sufficiency for regulated expression .

  2. Osteoclast Metabolism: TNFSF11 induces glutamine-dependent energy metabolism in osteoclasts, influencing bone resorption .

  3. Gender-Specific Effects: Mammary gland development during pregnancy requires TNFSF11 signaling .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered solution of phosphate-buffered saline (PBS), pH 7.4.
Form
Liquid or Lyophilized powder
Lead Time
Generally, we can ship products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timelines.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please contact us in advance as additional fees will apply.
Shelf Life
The shelf life of our products depends on various factors, including storage conditions, buffer composition, storage temperature, and the inherent stability of the protein itself.
Generally, liquid forms have a shelf life of 6 months at -20°C/-80°C, while lyophilized forms have a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
N-terminal 6xHis-tagged
Synonyms
Tnfsf11; Opgl; Rankl; Trance; Tumor necrosis factor ligand superfamily member 11; Osteoclast differentiation factor; ODF; Osteoprotegerin ligand; OPGL; Receptor activator of nuclear factor kappa-B ligand; RANKL; TNF-related activation-induced cytokine; TRANCE; CD antigen CD254) [Cleaved into: Tumor necrosis factor ligand superfamily member 11; membrane form; Tumor necrosis factor ligand superfamily member 11; soluble form]
Datasheet & Coa
Please contact us to get it.
Expression Region
73-316aa
Mol. Weight
28.5 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Cancer
Source
Mammalian cell
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Tumor necrosis factor ligand superfamily member 11 (TNFSF11), also known as RANKL, is a cytokine that plays a critical role in bone remodeling and immune regulation. It binds to two receptors: TNFRSF11B/OPG and TNFRSF11A/RANK. RANKL promotes osteoclast differentiation and activation, leading to bone resorption. It also enhances the ability of dendritic cells to stimulate naive T-cell proliferation, suggesting a role in regulating T-cell-dependent immune responses. Moreover, RANKL may contribute to enhanced bone resorption observed in humoral hypercalcemia of malignancy. RANKL induces osteoclastogenesis by activating multiple signaling pathways in osteoclast precursor cells, particularly by inducing sustained oscillations in intracellular calcium (Ca2+) levels. This activation triggers the translocation of NFATC1 to the nucleus, which in turn induces osteoclast-specific gene transcription, leading to osteoclast differentiation. During this process, RANKL activates CREB1 and mitochondrial ROS generation in a TMEM64 and ATP2A2-dependent manner, which are essential for proper osteoclast generation.
Gene References Into Functions
  1. These findings demonstrate that the membrane-bound form of RANKL is sufficient for most of its functions but that the soluble form does contribute to physiological bone remodeling in adult mice. PMID: 30046091
  2. At the molecular level, we confirmed, for the first time, that RES upregulated FoxO1 transcriptional activity by inhibiting the PI3K/AKT signaling pathway, which promoted resistance to oxidative damage and restrained osteoclastogenesis. Inhibition of the PI3K/AKT signaling pathway can be induced by RANKL. PMID: 29115382
  3. Deletion of the RANKL D5 enhancer delays the progression of atherosclerotic plaque development and plaque calcification in hypercholesterolemic mice. PMID: 28419519
  4. These results suggest that A2BAR stimulation inhibits the activation of ERK1/2, p38, and NF-kappaB by RANKL, which suppresses the induction of osteoclast marker genes, thus contributing to the decrease in osteoclast cell-cell fusion and bone resorption activity. PMID: 29047264
  5. Compressive force induced the differentiation of RAW264.7 cells through an increase in RANK expression and a decrease in LGR4 expression. PMID: 29572179
  6. miR-145 expression was inhibited in RANKL-induced osteoclastogenesis. PMID: 29577879
  7. LPS increased mRNA and protein expressions of IL-6 and RANKL on day 14. PMID: 28637991
  8. hBD-1 potentiates the induction of in vitro osteoclastogenesis by RANKL via enhanced phosphorylation of the p44/42 MAPKs. PMID: 28709835
  9. In conclusion, these results suggest that linarin has anti-osteoclastic effects and may serve as potential modulatory agents for the prevention and treatment of bone loss-associated diseases. PMID: 29269297
  10. Rankl(-/-) bone marrow-mesenchymal stromal cells displayed reduced clonogenicity and osteogenic capacity. PMID: 28100034
  11. In this study, the authors identified by gene expression profiling that microgravity induces high levels of TRAIL expression in murine preosteoclast cells in the absence of RANKL stimulation compared to ground-based cultures. PMID: 27142480
  12. These results suggest that LOX has the ability to induce RANKL expression on stromal cells; however, it fails to substitute for RANKL in osteoclastogenesis. PMID: 27606829
  13. Results demonstrated that picroside II strongly inhibited RANKL-induced osteoclast formation when added during the early stage of BMMs cultures, suggesting that it acts on osteoclast precursors to inhibit RANKL/RANK signaling. PMID: 28464271
  14. Loss of BMP signaling specifically in osteocytes dramatically increases bone mass presumably through simultaneous inhibition of RANKL and SOST, leading to osteoclast inhibition and Wnt activation together. PMID: 27402532
  15. The potentiation of RANKL-induced CTX release by dexamethasone was significantly less in bone marrow macrophage cells from mice with conditional knockout of the osteoclastic glucocorticoid receptor and completely absent in cells from GR(dim) mice, which carry a point mutation in one dimerizing interface of the GC receptor. PMID: 27596806
  16. Findings demonstrate that mTORC1 activation-stimulated RANKL expression in B cells is sufficient to induce bone loss and osteoporosis. The study also established a link between mTORC1 and the RANKL/OPG axis via negative regulation of beta-catenin. PMID: 26825871
  17. Dihydroartemisinin inhibited RANKL-induced NF-kappaB and NFAT activity. PMID: 26684711
  18. Cyanidin chloride is capable of inhibiting osteoclast formation, hydroxyapatite resorption, and RANKL-induced signal pathways in vitro and ovariectomy-induced bone loss in vivo. PMID: 28771720
  19. Together these data indicate that osteocyte apoptosis plays a central and controlling role in triggering osteocyte RANKL production and the activation of new resorption leading to bone loss in disuse. PMID: 26852281
  20. Data (including data from studies using knockout mice) suggest that RANKL enhances TNF-induced osteoclast formation from precursor spleen cells and enhances bone resorption independently of Traf6 by degrading Traf3, a known inhibitor of osteoclastogenesis. (RANKL = osteoclast differentiation factor; TNF = tumor necrosis factor; Traf = TNF receptor-associated factor) PMID: 28438834
  21. Results suggest that the MAPK signaling pathway is necessary for receptor activator of nuclear factor kappa-B ligand (RANKL) expression in bone marrow stromal cells. PMID: 28161637
  22. Endocortical resorption is driven by reduced osteoprotegerin rather than elevated RANKL expression. PMID: 27460899
  23. The results showed that AG490 inhibited (p)-JAK2 and RANKL expression. PMID: 28278513
  24. RANKL plays a supportive role in the RANKL-dependent differentiation. PMID: 27413168
  25. Increased expression of RANKL in heterogeneous bone marrow cells provoked bone destruction during Group A Streptococcus infection. PMID: 26894505
  26. Beta-lapachone inhibits RANKL-induced osteoclastogenesis and could be considered a potent inhibitor of RANKL-mediated bone diseases. PMID: 27913299
  27. This study demonstrates that PEMF is beneficial against RANKL-dependent osteoclastic differentiation in RAW264.7 cells in vitro via inhibiting the Ca2+-calcineurin-NFATc1 signaling pathway. PMID: 27856256
  28. These results demonstrate that RANKL expressed by osteocytes is required for the bone loss as well as the increase in B cell number caused by estrogen deficiency. PMID: 27733688
  29. In conclusion, the present study demonstrated that EA can suppress osteoclastogenesis in vitro. Moreover, we clarified that these inhibitory effects of EA occur through suppression of NF-kappaB and ERK activation. Therefore, EA may be a potential agent in the treatment of osteoclast-related diseases such as osteoporosis. PMID: 27349866
  30. MDH1 expression was induced by receptor activator of nuclear factor kappa-B ligand (RANKL) treatment. PMID: 27179783
  31. miR-338-3p may play a significant role in glucocorticoid-induced osteoclast differentiation and function by targeting RANKL in osteoclasts. PMID: 27706599
  32. The protein and RNA levels of RANKL-induced cFos and nuclear factor of activated T-cell cytoplasmic 1 were suppressed by centipedegrass extract (CGE). These results indicated that CGE may serve as a useful drug in the prevention of bone loss. PMID: 27035226
  33. Lipopolysaccharides significantly up-regulated RANKL expression and activated the ERK1/2 pathway to induce IL-6 mRNA expression and protein synthesis in MLO-Y4 cells. PMID: 27778412
  34. This study shows that OPG constitutes an early biomarker with in experimental model of severe malaria. PMID: 26766771
  35. Osteoprotegerin plays a role in preserving myocardial structure and function with ageing through a reduction in apoptosis and preservation of the matrix structure. PMID: 26825553
  36. These results indicate that activation of Panx1 and P2X7 R are required for apoptotic osteocytes in fatigued bone to trigger RANKL production in neighboring bystander osteocytes and implicate ATP as an essential signal mediating this process. PMID: 26553756

Show More

Hide All

Database Links
Involvement In Disease
Deficiency in Tnfsf11 results in failure to form lobulo-alveolar mammary structures during pregnancy, resulting in death of newborns. Trance-deficient mice show severe osteopetrosis, with no osteoclasts, marrow spaces, or tooth eruption, and exhibit profound growth retardation at several skeletal sites, including the limbs, skull, and vertebrae and have marked chondrodysplasia, with thick, irregular growth plates and a relative increase in hypertrophic chondrocytes.
Protein Families
Tumor necrosis factor family
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type II membrane protein.; [Isoform 2]: Cell membrane; Single-pass type II membrane protein.; [Isoform 3]: Cytoplasm.; [Tumor necrosis factor ligand superfamily member 11, soluble form]: Secreted.
Tissue Specificity
Highly expressed in thymus and lymph nodes, but not in non-lymphoid tissues and is abundantly expressed in T-cells but not in B-cells. A high level expression is also seen in the trabecular bone and lung.

Q&A

What is the molecular structure of mouse TNFSF11?

Mouse TNFSF11 is a type II transmembrane protein comprising 316 amino acids, with a predicted cytoplasmic domain of 48 amino acids and an extracellular domain of 247 amino acids. The extracellular domain contains two potential N-linked glycosylation sites. The protein functions as a homotrimer, which is essential for receptor binding. Mouse and human TNFSF11 share approximately 85% amino acid identity . The protein is expressed in two forms: a membrane-bound form and a soluble form, both of which are biologically active .

Which tissues and cells primarily express TNFSF11?

TNFSF11 is primarily expressed in T cells and T cell-rich organs such as the thymus and lymph nodes . Additionally, osteoblasts and osteocytes are major producers of TNFSF11 in the skeletal system. Activated T-cells also contribute significantly to TNFSF11 production, particularly in inflammatory conditions . This tissue-specific expression pattern reflects the protein's diverse roles in both the immune and skeletal systems.

How is TNFSF11 gene expression regulated?

The expression of TNFSF11 is controlled by several factors, including vitamin D, parathyroid hormone, and various cytokines . In humans, the TNFSF11 gene is mapped to chromosome 13q14 . Epigenetic regulation through DNA methylation of the TNFSF11 promoter region has also been identified as an important regulatory mechanism, with hypomethylation associated with increased expression in certain pathological conditions . The promoter regulatory region spans from −260 to +615 bp of the transcription start site (TSS) of isoform 1 .

What is the RANK/RANKL/OPG axis and how does it function?

The RANK/RANKL/OPG axis is a crucial regulatory system for bone remodeling. RANKL (TNFSF11) binds to its receptor RANK (expressed on osteoclast precursor cells), initiating a series of events that results in osteoclast differentiation and activation. Osteoprotegerin (OPG), synthesized by osteoblasts and other cells, acts as a decoy receptor that binds to RANKL, preventing its interaction with RANK and thereby inhibiting osteoclastogenesis. The proper balance between RANKL and OPG levels is essential for maintaining bone homeostasis, and imbalances in this system have been implicated in the pathogenesis of osteoporosis and other bone disorders .

What are the key molecular signaling pathways activated by TNFSF11?

When TNFSF11 binds to RANK on target cells, it triggers several intracellular signaling cascades. This binding initiates the activation of NF-κB, MAPKs, and calcium signaling pathways. At the molecular level, the RANKL-RANK interaction involves recruitment of TRAF6, which leads to a series of phosphorylation and ubiquitination processes. These molecular events ultimately result in the activation of transcription factors that control osteoclast differentiation . During osteoclast differentiation, TNFSF11 induces activation of CREB1 and mitochondrial ROS generation in a TMEM64 and ATP2A2-dependent manner, which is necessary for proper osteoclast development .

Beyond bone metabolism, what other biological functions does TNFSF11 influence?

TNFSF11 has multiple functions beyond bone metabolism. It enhances T cell growth and dendritic cell function, augmenting the ability of dendritic cells to stimulate naive T-cell proliferation. TNFSF11 may be an important regulator of interactions between T-cells and dendritic cells, playing a role in the regulation of T-cell-dependent immune responses. Additionally, it is involved in lymph node organogenesis and may play an important role in enhanced bone-resorption in humoral hypercalcemia of malignancy . These diverse functions highlight TNFSF11's role as a pleiotropic cytokine affecting multiple physiological systems.

How can recombinant mouse TNFSF11 be used in osteoclast differentiation assays?

Recombinant mouse TNFSF11 can effectively induce osteoclast differentiation of RAW 264.7 mouse monocyte/macrophage cell lines, with an ED50 (effective dose for 50% response) of 0.5-2 ng/mL . For experimental protocols, researchers should:

  • Culture RAW 264.7 cells in appropriate medium (typically DMEM with 10% FBS)

  • Seed cells at optimal density (approximately 1-2 × 10⁴ cells/well in 96-well plates)

  • Add recombinant mouse TNFSF11 at concentrations ranging from 0.1-100 ng/mL (include a dose-response curve)

  • Incubate for 4-7 days, replacing medium and TNFSF11 every 2-3 days

  • Assess osteoclast formation by TRAP (tartrate-resistant acid phosphatase) staining and counting multinucleated TRAP-positive cells

This methodology allows for quantitative assessment of osteoclastogenic potential and can be adapted to test inhibitors or enhancers of this process.

What methods are recommended for analyzing TNFSF11 methylation patterns?

To analyze TNFSF11 promoter methylation patterns, a recommended methodology involves bisulfite conversion followed by methylation-specific PCR (MSP). The protocol should include:

  • DNA extraction and bisulfite conversion (which converts unmethylated cytosines to uracils while leaving methylated cytosines unchanged)

  • PCR amplification using primers specific for methylated and unmethylated DNA in the TNFSF11 promoter region (spanning from −260 to +615 bp of the TSS)

  • Internal normalization with optimized primers for known genomic regions (e.g., TSH2B for methylated DNA and GAPDH for unmethylated DNA)

  • Use of positive controls (human bisulfite-converted methylated and unmethylated DNA controls)

  • Quantification using real-time PCR and calculation of methylation percentages

The ΔΔCq method can be used to assess the difference in relative methylation levels between samples, with reference genes for normalization . This approach provides insights into epigenetic regulation of TNFSF11 expression.

What quality control parameters should be assessed for recombinant mouse TNFSF11 in research applications?

When working with recombinant mouse TNFSF11, researchers should evaluate several quality control parameters:

  • Purity assessment: Use SDS-PAGE under reducing conditions (should show a single band at approximately 19 kDa)

  • Biological activity: Verify through osteoclast differentiation assays using RAW 264.7 cells (ED50 of 0.5-2 ng/mL)

  • Endotoxin levels: Should be tested and maintained below acceptable thresholds for cell culture work

  • Protein concentration: Accurately determine using established protein quantification methods

  • Stability testing: Assess activity retention after reconstitution under recommended storage conditions

These parameters ensure experimental reproducibility and reliability when working with recombinant TNFSF11.

How does the rs1021188 polymorphism influence TNFSF11 function and disease susceptibility?

The rs1021188 polymorphism has been associated with altered TNFSF11 expression and disease susceptibility. Research has shown:

  • This polymorphism is located in the regulatory region of the TNFSF11 gene

  • It has been linked to otosclerosis (OTSC), with significant differences in genotype frequencies between patients and healthy controls

  • The variant may affect transcription factor binding sites and alter gene expression levels

  • There appears to be an independent effect of the rs1021188 polymorphism and DNA hypomethylation of the TNFSF11 promoter in OTSC

To study this association, researchers should consider genotyping using direct counting methods, Hardy-Weinberg Equilibrium testing for genotype frequency deviation in control groups, and statistical comparison of genotypes between case and control groups using Chi-square tests .

What is the relationship between TNFSF11 expression, genetic variation, and bone disorders?

TNFSF11 plays a critical role in bone metabolism, and alterations in its expression or function are associated with several bone disorders:

  • Osteoporosis: Imbalances in the RANK/RANKL/OPG axis contribute to pathological bone loss

  • Otosclerosis (OTSC): Associated with rs1021188 polymorphism and DNA hypomethylation of the TNFSF11 promoter

  • Osteopetrosis: TNFSF11 deficiency has been linked to Autosomal Recessive Osteopetrosis 2 and Autosomal Recessive Malignant Osteopetrosis

Research approaches should include genotype-phenotype correlation studies, functional analyses of genetic variants, and investigation of epigenetic modifications affecting TNFSF11 expression in disease states.

How can regulatory regions of the TNFSF11 gene be analyzed for evolutionary conservation?

Analysis of evolutionary conservation of TNFSF11 regulatory regions, particularly CpG islands, provides insights into functionally important elements. A methodological approach includes:

  • Utilize the University of California, Santa Cruz Genome Browser interface (GRCh37/hg19 assembly)

  • Perform multiple alignments across 100 vertebrate species

  • Measure evolutionary conservation using phastCons and phyloP programs

  • Classify CpG-rich regions according to their evolutionary dynamics using parameter-rich evolutionary models and clustering analysis

  • Evaluate chromatin non-condensed DNaseI Hypersensitivity sites in multiple cell types

What approaches can be used to study the epigenetic regulation of TNFSF11 in different disease models?

To investigate epigenetic regulation of TNFSF11 across disease models, researchers should employ:

  • Bisulfite sequencing: For comprehensive DNA methylation profiling of the TNFSF11 promoter

  • Chromatin immunoprecipitation (ChIP): To identify histone modifications and transcription factor binding at the TNFSF11 locus

  • CRISPR-based epigenome editing: To causally test the impact of specific epigenetic modifications

  • Single-cell approaches: To characterize cell-type specific epigenetic states

  • Integrative bioinformatics: To correlate epigenetic patterns with gene expression and disease phenotypes

Comparing methylation patterns between disease and healthy samples requires statistical evaluation using non-parametric tests (e.g., Mann-Whitney) and potentially multivariate analysis with hierarchical clustering .

What are the challenges in developing highly specific inhibitors or modulators of the RANKL-RANK interaction?

Developing specific modulators of the RANKL-RANK interaction presents several challenges:

  • Structural considerations: The protein functions as a homotrimer, requiring modulators that can interfere with trimerization or receptor clustering

  • Specificity: Ensuring that modulators target only the RANKL-RANK interaction without affecting other TNF family members

  • Tissue-specific targeting: Developing approaches that can target specific tissues (e.g., bone vs. immune system) to limit off-target effects

  • Functional redundancy: Addressing potential compensatory mechanisms in the TNF superfamily

  • Dosing and pharmacokinetics: Determining optimal dosing regimens that effectively modulate the pathway without complete inhibition

Research approaches should include structure-based drug design, high-throughput screening of compound libraries, and development of tissue-specific delivery systems.

What are emerging areas of TNFSF11 research beyond traditional bone and immune applications?

Emerging research directions for TNFSF11 include:

  • Role in neuroinflammation and neurodegenerative disorders

  • Involvement in metabolic diseases and adipose tissue biology

  • Functions in cancer progression, particularly in tumor-induced bone disease

  • Potential as a biomarker for disease progression in various conditions

  • Interactions with the microbiome and subsequent effects on bone and immune homeostasis

These expanding areas highlight the pleiotropic nature of TNFSF11 and suggest new therapeutic targets and diagnostic approaches.

How can systems biology approaches enhance our understanding of TNFSF11 in health and disease?

Systems biology approaches offer powerful tools for understanding TNFSF11's complex roles:

  • Network analysis: Mapping TNFSF11 interactions within broader signaling networks

  • Multi-omics integration: Combining genomics, transcriptomics, proteomics, and metabolomics data

  • Mathematical modeling: Developing predictive models of RANKL-RANK-OPG dynamics

  • In silico screening: Computational identification of potential modulators

  • Pathway enrichment analysis: Identifying biological processes affected by TNFSF11 dysregulation

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.