Recombinant Human Tumor necrosis factor receptor superfamily member 11A protein (TNFRSF11A), partial (Active)

Shipped with Ice Packs
In Stock

Description

Biological Function and Activity

TNFRSF11A is essential for osteoclastogenesis and bone remodeling. Its partial recombinant forms are used to study:

Core Mechanisms

  1. RANKL Binding:

    • Recombinant TNFRSF11A binds RANKL, blocking its interaction with native receptors .

    • Active variants inhibit osteoclast differentiation in vitro (IC₅₀: 0.005–0.035 µg/mL) .

  2. Signaling Pathways:

    • Activates NF-κB and JNK pathways via TRAF proteins .

    • Modulates T-cell–dendritic cell interactions and lymph node development .

Disease Associations

DiseaseMechanismSources
Paget DiseaseDuplication mutations in TNFRSF11A hyperactivate osteoclast signaling .
Familial Expansile OsteolysisOveractive RANK leads to excessive bone resorption .
AsthmaTNFSF11/TNFRSF11A axis amplifies airway remodeling via STAT3/TGFβ1 .

Key Applications

Use CaseMethodologyOutcome
Osteoclast InhibitionCo-culture with RANKL and precursor cells (e.g., RAW264.7 macrophages) .Reduced TRAP+ osteoclast formation by >50% at 0.1 µg/mL .
Asthma ModelingHDM-induced airway remodeling in Tnfrsf11a +/− mice or HBE cells .↓ STAT3 phosphorylation and ↓ collagen deposition in bronchi .
Therapeutic ScreeningDenosumab (anti-TNFSF11) treatment in humanized mice .Attenuated airway hyperresponsiveness and fibrosis .

Mechanistic Insights from Recent Studies (2024)

  • TNFSF11/TNFRSF11A Axis in Asthma:

    • Upregulated in bronchial epithelia of asthma patients .

    • Silencing TNFSF11 or TNFRSF11A via shRNA reduces TGFβ1/STAT3 signaling and collagen synthesis .

    • Denosumab (anti-TNFSF11) reverses HDM-induced remodeling in humanized mice .

Manufacturing Process

StepDetailsSources
CloningSynthetic DNA encoding aa 28–202 or 29–213 inserted into plasmids .
ExpressionTransfected into HEK-293 or E. coli cells for high-yield production .
PurificationAffinity chromatography (His/Strep tags) or size-exclusion chromatography .
ValidationSDS-PAGE, Western blot, and functional assays (e.g., ELISA for RANKL binding) .

Quality Metrics

ParameterSpecificationSources
Endotoxin<0.01 EU/μg (LAL test) .
Storage-80°C for GST-tagged proteins; -20°C or 4°C for Fc-tagged variants .

Targeted Therapies

AgentMechanismApplication
DenosumabMonoclonal antibody against TNFSF11, blocking RANKL-TNFRSF11A interaction .Osteoporosis, giant cell tumor, and asthma .
Osteoprotegerin (OPG)Soluble decoy receptor for RANKL, inhibiting osteoclastogenesis .Bone metastasis and Paget disease .

Ongoing Research Directions

  1. Asthma Therapy:

    • Blocking TNFSF11/TNFRSF11A axis to reduce airway fibrosis .

  2. Bone Disorders:

    • Gene therapy for familial expansile osteolysis or Paget disease .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 20 mM Tris-HCl, pH 8.0, 150mM NaCl
Description

The Recombinant Human Tumor necrosis factor receptor superfamily member 11A protein (TNFRSF11A) is a partial-length protein encompassing amino acids 29-202 of the human TNFRSF11A. This protein was produced through the expression of a DNA fragment encoding this region in E. coli. Rigorous validation via SDS-PAGE and HPLC analyses confirmed a purity exceeding 90%. Furthermore, its biological activity was verified through its interaction with sRANK Ligand. The endotoxin level of this TNFRSF11A is below 1.0 EU/µg as determined by the LAL method. This recombinant TNFRSF11A protein is readily available, eliminating any waiting time for preparation. Custom tagged proteins are also offered upon request. This recombinant TNFRSF11A protein can be utilized for the development of specific antibodies or in research focusing on TNFRSF11A-associated cancers.

TNFRSF11A, also known as RANK, serves as the receptor for RANKL. The interaction between RANK and RANKL plays a critical role in a multitude of biological processes, including bone resorption and remodeling, bone marrow formation, development of the thymus and lymph nodes, intestinal immunity, inflammatory responses within the skin and central nervous system (CNS), tumorigenesis, and metastasis. Mutations within RANK or RANKL have been linked to the pathogenesis of bone diseases and certain types of cancer.

Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
It is recommended to briefly centrifuge this vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to achieve a concentration between 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our default final concentration of glycerol is 50% and can be used as a reference.
Shelf Life
The shelf life is influenced by various factors including storage conditions, buffer composition, storage temperature, and the inherent stability of the protein. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C, while the lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
CD 265; CD265; FEO; LOH18CR1; Loss of heterozygosity 18 chromosomal region 1; mRANK; ODFR; OFE; OPTB7; Osteoclast differentiation factor receptor; OSTS; Paget disease of bone 2; PDB 2; PDB2; RANK; Receptor activator of NF KB; Receptor activator of NF-KB; receptor activator of nuclear factor kappa B; TNF receptor superfamily member 11a; TNFRSF11A; TNR11_HUMAN; TRANCER; Tumor necrosis factor receptor superfamily member 11a NFKB activator; Tumor necrosis factor receptor superfamily member 11a activator of NFKB; Tumor necrosis factor receptor superfamily member 11A
Datasheet & Coa
Please contact us to get it.
Expression Region
29-202aa
Mol. Weight
19.1 kDa
Protein Length
Partial
Purity
>98% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function

This receptor for TNFSF11/RANKL/TRANCE/OPGL is essential for RANKL-mediated osteoclastogenesis. It plays a significant role in regulating interactions between T-cells and dendritic cells.

Gene References Into Functions
  1. Reduced miR-144-3p expression in serum and bone mediates osteoporosis pathogenesis by targeting RANK. PMID: 29334613
  2. Findings indicate that C/EBPalpha is a stronger inducer of osteoclast differentiation than c-Fos, partly via C/EBPalpha regulation by the RANK (535)IVVY(538) motif PMID: 29122885
  3. gammadelta T cells suppressed iDCs osteoclastogenesis by downregulation of the RANK/cFos/ATP6V0D2 signaling pathway. PMID: 30066839
  4. RANK protein expression increased from normal to malignant endometrium, and the expression level was related with tumor grade but not with stage or the age of subjects in endometrial cancer. PMID: 29932437
  5. Study identified the second disease gene for DOS. TNFRSF11A isoforms may have the different roles in skeletal development and metabolism PMID: 29568001
  6. The mRNA expression of RANK was highest in prostate tumour tissue from patients with bone metastases as compared to BPH or locally confined tumours, also shown in clinical subgroups distinguished by Gleason Score or PSA level. PMID: 29204705
  7. RANK 575C>T polymorphisms did not show any statistically significant differences between the study groups (Osteoporosis and Osteopenia) and Postmenopausal women. PMID: 27304650
  8. For the RANK gene, the AGTGC haplotype was associated with the lowest risk of presenting chronic joint pain in individuals without TMD (P=0.03). This study supports the hypothesis that changes in the OPG and RANK genes influence the presence of chronic joint pain in individuals with and without TMD. PMID: 28464982
  9. In this study, whole exome sequencing (WES) was successfully used in six patients with malignant infantile osteopetrosis (MIOP) and identified mutations in four MIOP-related genes (CLCN7, TCIRG1, SNX10, and TNFRSF11A). PMID: 27187610
  10. RANK is increased in hormone receptor negative and basal breast cancer, and correlates with worse recurrence-free survival and risk of bone metastasis. PMID: 28577080
  11. RANK SNP rs34945627 has a high allelic frequency in patients with breast cancer and Bone metastases, and is associated with decreased disease-free survival and Overall Survival. PMID: 27191503
  12. RANK rewires energy homeostasis in human lung cancer cells and promotes expansion of lung cancer stem-like cells. PMID: 29118048
  13. EGFR and RANK combinatorial in vitro analyses revealed a significant upregulation of AKT and ERK signaling after EGF stimulation in cell lines and also an increase of breast cancer cell invasiveness. PMID: 29025596
  14. In histologically normal tissue of BRCA1-mutation carriers and showed that RANK(+) cells are highly proliferative, have grossly aberrant DNA repair and bear a molecular signature similar to that of basal-like breast cancer. PMID: 27322743
  15. Vav3 is a novel TRAF6 interaction partner that functions in the activation of cooperative signaling between T6BSs and the IVVY motif in the RANK signaling complex. PMID: 27507811
  16. RANK and CCR6 expressed on monocytes may be novel targets for the regulation of bone resorption in rheumatoid arthritis and osteoporosis. PMID: 27822475
  17. High RANK expression is associated with endometrial metastasis. PMID: 26734994
  18. Study showed that endogenous RANK expression changes might influence prostate cancer cell behavior since reduced RANK expression resulted in significantly increased PC-3 cell proliferation and adhesion. PMID: 26977008
  19. genetic variation associated with hypertension in Chinese women PMID: 25810067
  20. we could not identify any association between external apical root resorption and two SNPs, rs1805034 from TNFRSF11A (encoding RANK) and rs3102735 from TNFRSF11B (encoding OPG). PMID: 24118270
  21. Results suggest that Cbl-b improves the prognosis of RANK-expressing breast cancer patients by inhibiting RANKL-induced breast cancer cell migration and metastasis. PMID: 26087197
  22. Based on our findings, the functional SNP RANK rs1805034 T>C may be an indicator for individual susceptibility to GCA. PMID: 26451891
  23. Functional polymorphisms RANK rs1805034 T>C may be an indicator for individual susceptibility to esophageal squamous cell carcinoma. PMID: 25019155
  24. These findings indicate that epidermal leukocytes gradually acquire RANK during gestation - a phenomenon previously observed also for other markers on LCs in prenatal human skin. PMID: 25722033
  25. this study is the first to identify RANK overexpression as a novel esophageal cancer marker in both Kazakh and Han ethnic esophageal squamous cell carcinoma patients. PMID: 25973136
  26. RANK/OPG ratio of expression in primary ccRCC is associated with BM. PMID: 26528707
  27. Three single nucleotide polymorphisms of TNFRSF11A (rs4500848, rs6567270 and rs1805034) are associated with Age at menarche and Age at natural menopause in Chinese women. PMID: 25884698
  28. Microcomputed tomography analysis demonstrated that the mice treated with rhRANK exhibited an increased bone volume and structure model index, and decreased trabecular spacing compared with those treated with rhOPG-Fc. PMID: 25738879
  29. results indicate that the RANK IVVY motif cooperates with the TRAF-binding motifs to promote osteoclastogenesis, which provides novel insights into the molecular mechanism of RANK signaling in osteoclastogenesis. PMID: 26276390
  30. Response to sRANKL in normal and tumor cells suggests a role for RANK/ERK-mediated signaling in normal osteoblasts chemotactic migration during bone remodeling that is altered or lost during osteosarcoma tumorigenesis. PMID: 25893522
  31. Higher RANK expression in the primary breast tumor is associated with a higher sensitivity to chemotherapy, but also a higher risk of relapse and death. PMID: 24737168
  32. Changes in bone markers, OPG, sRANKL and/or the OPG/sRANKL ratio exhibited by girls with Anorexia nervosa have been found to be associated with changes in the levels of the selected adipose tissue hormones. PMID: 24549600
  33. Genetic polymorphism in TNFRSF11A influences bone mineral density in post-menopausal women. PMID: 25138264
  34. Single nucleotide polymorphisms of RANK and PTGS1 show genetic associations with osteoproliferative changes in ankylosing spondylitis. PMID: 24651623
  35. he top hit rs17069906 (p = 5.6 e-10) is located within the genomic region of RANK, recently demonstrated to be an important player in the adaptive recovery response in podocytes and suggested as a promising therapeutic target in glomerular diseases. PMID: 25478860
  36. This review study has focused on the association of RANKL-RANK-OPG pathway in the pathogenesis and progression of giant cell tumor of bone as well as discussed the possible therapeutic strategies by targeting this pathway PMID: 25618600
  37. The urinary mRNA of RANK might be used to differentiate histologic subtypes of glomerulonephritis, particularly between minimal change disease and membranous nephropathy. PMID: 25171769
  38. RANKL and IL-6 mediate direct paracrine-autocrine signaling between cells of the osteoblast lineage and cancer cells. PMID: 24676805
  39. PGRN and PIRO form a new regulatory axis in osteoclastogenesis that is included in RANK signaling in cell fusion and OC resorption of osteoclastogenesis PMID: 25406312
  40. we identified RANK expression as a negative prognostic factor regarding disease-free survival in osteosarcoma PMID: 24842377
  41. High RANK protein expression is associated with breast cancer. PMID: 25111682
  42. RANKL, either derived from the prostate tumor or from the host, plays a key role in cancer bone metastasis. PMID: 24478054
  43. The involvement of TNFRSF11A in hereditary recurrent fever highlights the key role of this receptor in innate immunity. PMID: 24891336
  44. Mechanistic studies showed that IL-10 downregulated RANK expression in monocytes and thus, inhibited RANKL-induced OC formation PMID: 24340030
  45. silencing of miR-503 using a specific antagomir in ovariectomy (OVX) mice increased RANK protein expression, promoted bone resorption PMID: 23821519
  46. The genotypes, combined genotypes and allele frequencies of C421T and C575T polymorphisms of the RANK gene have not been found to be associated with bone mineral density in Turkish women. PMID: 23553199
  47. RANK signaling interferes with mammary cell commitment, contributing to breast carcinogenesis. PMID: 23766243
  48. High RANK expression is associated with Avascular Necrosis of Femur Head. PMID: 24200492
  49. The atrial expression of RANK (and RANKL/osteoprotegerin ratio) was higher in normal controls compared to persistent atrial fibrillation patients. PMID: 22178057
  50. One mechanism of RANK inhibition by 1,25(OH)2D3 is down-regulation of the M-CSF receptor c-Fms, which is required for the expression of RANK. PMID: 23116709

Show More

Hide All

Database Links

HGNC: 11908

OMIM: 174810

KEGG: hsa:8792

STRING: 9606.ENSP00000465500

UniGene: Hs.204044

Involvement In Disease
Familial expansile osteolysis (FEO); Paget disease of bone 2, early-onset (PDB2); Osteopetrosis, autosomal recessive 7 (OPTB7)
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein.; [Isoform RANK-e5a]: Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Ubiquitous expression with high levels in skeletal muscle, thymus, liver, colon, small intestine and adrenal gland.

Q&A

What is TNFRSF11A and what are its primary physiological functions?

TNFRSF11A (TNF receptor superfamily member 11a), commonly known as RANK (Receptor Activator of Nuclear Factor κB), is a type I transmembrane protein belonging to the tumor necrosis factor receptor family. The full-length human RANK contains a 184 amino acid extracellular domain and a 383 amino acid cytoplasmic domain, with two potential N-linked glycosylation sites in its extracellular region . TNFRSF11A serves several critical physiological functions, primarily as an essential mediator for osteoclast differentiation and lymph node development .

The protein functions as a receptor that interacts with various TRAF (TNF Receptor Associated Factor) family proteins, through which it induces the activation of key signaling pathways, particularly NF-kappa B and MAPK8/JNK . These signaling cascades are crucial for several biological processes including:

  • Regulation of interactions between T cells and dendritic cells in the immune system

  • Mediation of osteoclast formation and activation in bone remodeling

  • Development and organization of lymph nodes

  • Enhancement of T cell growth and dendritic cell function

The protein is widely expressed in human tissues, with highest expression observed in skeletal muscle, thymus, liver, colon, small intestine, and adrenal gland .

How is TNFRSF11A expression regulated in different cell types?

Research with neutrophils has demonstrated that synovial fluid from rheumatoid arthritis (RA) patients can induce the expression of RANK, suggesting environmental and inflammatory factors may regulate its expression . This induction appears to be part of a broader regulatory network involving RANK, RANKL (RANK ligand), and OPG (osteoprotegerin), which collectively control osteoclast differentiation and activity.

TNFRSF11A expression is also subject to alternative splicing, with several splice variants identified in breast cancer cell lines, including TNFRSF11A_delta 9 (RANK-a), TNFRSF11A_delta 8,9 (RANK-b), and TNFRSF11A_delta 7,8,9 (RANK-c) . These alternative splice variants may have distinct functional properties and could be regulated differently across tissue types.

What cellular signaling pathways are activated by TNFRSF11A?

TNFRSF11A activates multiple signaling pathways critical for cellular differentiation and function. The primary signaling mechanism involves the recruitment of TRAF family proteins to its cytoplasmic domain, which subsequently activates downstream pathways . The major signaling pathways include:

  • NF-κB (Nuclear Factor-kappa B) Pathway: TNFRSF11A stimulation leads to activation of both canonical and non-canonical NF-κB pathways, resulting in the translocation of transcription factors to the nucleus and expression of target genes involved in osteoclast differentiation, survival, and function .

  • MAPK (Mitogen-Activated Protein Kinase) Pathways: TNFRSF11A activates multiple MAPK pathways, particularly the c-Jun N-terminal kinase (JNK) pathway, which regulates cellular responses to stress, inflammatory cytokines, and growth factors .

  • PI3K/Akt Pathway: This pathway contributes to osteoclast survival and cytoskeletal rearrangements necessary for bone resorption.

  • NFATc1 (Nuclear Factor of Activated T cells c1) Pathway: TNFRSF11A signaling ultimately leads to the activation and amplification of NFATc1, a master transcription factor for osteoclastogenesis.

These signaling cascades work in concert to mediate various biological effects, including enhancement of T cell growth, dendritic cell function, induction of osteoclastogenesis, and lymph node organogenesis .

What is the genomic structure of TNFRSF11A and where is it located?

TNFRSF11A is located on the long arm of chromosome 18 at position 18q21.33 . The gene consists of 12 exons that encode the full protein . Within the genome, TNFRSF11A has been mapped between markers D18S383 and D18S51, specifically in the region of D18S60 between WI-9823 and SGC33768 .

The genomic sequence is represented by the reference sequence NC_000018.10 (62325310..62391288) on chromosome 18 . The full-length human TNFRSF11A cDNA encodes a type I transmembrane protein of 616 amino acids . The gene contains several polymorphic sites that have been identified, including variants in the 5'-UTR (-1G/A, 30T/C), coding regions (575C/T, 421C/T), and intronic regions (IVS6+79A/G) .

Understanding the genomic organization of TNFRSF11A is particularly important for mutation analysis in associated disorders and for designing gene-targeting strategies in research applications.

What methodologies are optimal for studying TNFRSF11A-mediated signaling in osteoclast differentiation?

The study of TNFRSF11A-mediated signaling in osteoclast differentiation requires a multi-faceted approach combining molecular, cellular, and biochemical techniques. Based on current research practices, the following methodological approaches are recommended:

  • Cell Culture Systems:

    • RAW 264.7 mouse monocyte/macrophage cell line is an established model for studying RANK-mediated osteoclastogenesis

    • Primary bone marrow macrophages (BMMs) isolated from mice provide a physiologically relevant system

    • Human peripheral blood mononuclear cells (PBMCs) can be used for translational studies

  • Osteoclast Differentiation Assays:

    • Treat RAW 264.7 cells with recombinant RANKL and M-CSF to induce osteoclast differentiation

    • Use TRAP (tartrate-resistant acid phosphatase) staining to identify mature osteoclasts

    • Measure bone resorption activity using synthetic bone substrates or bone slices

  • Signaling Pathway Analysis:

    • Western blotting to detect phosphorylation of key signaling molecules (NF-κB, MAPK, Akt)

    • Immunoprecipitation to study protein-protein interactions between RANK and TRAF adaptor proteins

    • Luciferase reporter assays to quantify NF-κB activation

    • Chromatin immunoprecipitation (ChIP) to analyze transcription factor binding to target gene promoters

  • Functional Manipulation:

    • Use TNFRSF11A agonistic antibodies (such as AF683) that can induce osteoclast differentiation with an ED50 of approximately 0.0750–1.50 μg/mL

    • Apply soluble RANK to block RANKL-induced biological activity

    • Utilize siRNA or CRISPR-Cas9 to knockdown or knockout TNFRSF11A expression

  • Expression Analysis:

    • RT-PCR and qPCR to measure TNFRSF11A mRNA expression and alternative splicing

    • Flow cytometry to analyze cell surface expression of RANK protein

    • Immunohistochemistry to visualize RANK expression in tissue sections

These methodologies can be combined to comprehensively study the role of TNFRSF11A in osteoclast differentiation, from receptor expression and activation to downstream signaling events and functional outcomes.

How do mutations in the signal peptide of TNFRSF11A affect protein function and contribute to bone pathologies?

Mutations in the signal peptide region of TNFRSF11A have profound effects on protein function and are directly linked to several bone pathologies. Research has identified specific tandem duplications in exon 1 of TNFRSF11A that affect the signal peptide of RANK and cause familial expansile osteolysis (FEO) and Paget's disease of bone (PDB) .

Molecular Mechanisms of Signal Peptide Mutations:

  • Altered Protein Processing: The identified mutations (84dup18 in FEO and 75dup27 in PDB) lead to perturbations in RANK expression levels and prevent normal cleavage of the signal peptide . This disruption in protein processing affects the proper localization and membrane insertion of the receptor.

  • Constitutive Activation: Both mutations cause an increase in RANK-mediated NF-κB signaling in vitro, consistent with an activating mutation . This leads to heightened downstream signaling even in the absence of RANKL stimulation.

  • Enhanced Osteoclastogenesis: The constitutive activation of RANK signaling results in increased osteoclast formation and activity, leading to excessive bone resorption and the characteristic osteolytic lesions observed in affected individuals.

Experimental Approaches to Study These Mutations:

  • Site-directed mutagenesis to introduce the duplications into wild-type TNFRSF11A cDNA

  • Expression of wild-type and mutant constructs in cell lines to assess differences in:

    • Protein localization (immunofluorescence microscopy)

    • Signal peptide cleavage (Western blot with antibodies against different protein domains)

    • NF-κB activation (luciferase reporter assays)

    • Osteoclast differentiation capacity (when expressed in osteoclast precursors)

  • Generation of knock-in mouse models carrying the human mutations to study the phenotypic consequences at the organismal level

Understanding the molecular consequences of these signal peptide mutations provides insights into both the normal function of TNFRSF11A and the pathogenesis of associated bone disorders, potentially identifying targets for therapeutic intervention.

What are the roles of TNFRSF11A splice variants in pathological conditions such as cancer?

TNFRSF11A undergoes alternative splicing, generating several variants with potentially distinct functional properties. Research has identified multiple splice variants in breast cancer cell lines, including TNFRSF11A_delta 9 (RANK-a), TNFRSF11A_delta 8,9 (RANK-b), and TNFRSF11A_delta 7,8,9 (RANK-c) . These splice variants may have significant implications in pathological conditions, particularly in cancer.

Methodological Approaches to Study Splice Variants:

  • Identification and Characterization:

    • RT-PCR with primers spanning multiple exons to detect alternative splicing events

    • Agarose gel electrophoresis to visualize different splice variants

    • Cloning and sequencing to confirm the identity of splice variants

    • qPCR with variant-specific primers to quantify relative expression levels

  • Expression Analysis in Pathological Conditions:

    • Comparative expression analysis in normal versus cancer cell lines

    • Assessment of splice variant expression in relation to clinical parameters such as tumor grade and proliferation marker Ki-67

    • Analysis in formaldehyde-fixed paraffin-embedded (FFPE) tissue samples from patients

Research Findings on TNFRSF11A Splice Variants in Cancer:

Research has shown that TNFRSF11A_delta 7,8,9 (RANK-c) expression varies across different breast cancer cell lines, with distinct expression patterns compared to the non-tumorigenic epithelial cell line MCF10A . This suggests a potential role for this splice variant in cancer development or progression.

In a panel of invasive ductal breast carcinoma samples, the relative expression levels of both wild-type RANK and the RANK-c variant showed relationships with tumor histological grade . This indicates that alterations in TNFRSF11A splicing may be associated with cancer aggressiveness.

Further investigation of these splice variants is warranted to understand:

  • Their structural and functional differences from full-length TNFRSF11A

  • Their impact on RANK-RANKL signaling and downstream pathways

  • Their potential as biomarkers or therapeutic targets in cancer

  • Their roles in other pathological conditions beyond breast cancer

How can TNFRSF11A-targeted approaches be developed for therapeutic applications in bone disorders?

Developing therapeutic approaches targeting TNFRSF11A requires understanding its structure-function relationships and its role in pathological conditions. Several strategies can be employed:

1. Antibody-Based Approaches:

  • Neutralizing antibodies against RANK to block interaction with RANKL

  • Agonistic antibodies (like AF683) that could potentially be modified to have partial agonist activity for fine-tuning RANK signaling

  • Engineering antibodies to target specific epitopes to modulate particular downstream pathways

2. Recombinant Protein Therapeutics:

  • Soluble RANK as a decoy receptor to compete with membrane-bound RANK for RANKL binding

  • Modified versions of RANK with enhanced binding affinity or altered signaling properties

  • Fusion proteins combining the RANKL-binding domain of RANK with other functional domains

3. Small Molecule Inhibitors:

  • Compounds targeting the intracellular signaling mediators of RANK (e.g., TRAF proteins)

  • Molecules that modulate specific downstream pathways like NF-κB or MAPK

  • Signal peptide-targeting compounds that could potentially correct the defects caused by the FEO and PDB mutations

4. Gene Therapy Approaches:

  • CRISPR-Cas9 gene editing to correct pathogenic mutations in TNFRSF11A

  • RNA interference to modulate expression of TNFRSF11A or its splice variants

  • Antisense oligonucleotides to alter splicing patterns in conditions where aberrant splicing contributes to pathology

5. Precision Medicine Strategies:

  • Genotyping patients for TNFRSF11A mutations or polymorphisms to guide therapeutic decisions

  • Monitoring expression of TNFRSF11A and its splice variants as biomarkers for disease progression or treatment response

  • Combining TNFRSF11A-targeted therapies with other agents for synergistic effects

Experimental Considerations:

  • In vitro osteoclast differentiation assays to assess efficacy

  • Bone resorption assays to determine functional impact

  • Animal models of FEO, PDB, or osteoporosis for in vivo validation

  • Safety assessment focusing on potential effects on immune function given RANK's role in dendritic cells and T cells

Development of these therapeutic approaches requires careful consideration of TNFRSF11A's physiological roles to minimize unwanted effects while maximizing therapeutic benefits in targeted bone disorders.

What are the optimal protocols for expressing and purifying recombinant TNFRSF11A protein for structural and functional studies?

Producing high-quality recombinant TNFRSF11A protein is essential for various research applications including structural studies, binding assays, and functional characterization. Based on established methodologies in protein biochemistry and the specific characteristics of TNFRSF11A, the following protocol considerations are recommended:

Expression System Selection:

  • Mammalian Expression Systems (Preferred for full-length or extracellular domain):

    • HEK293 or CHO cells are optimal for obtaining properly folded and glycosylated TNFRSF11A

    • These systems ensure correct formation of disulfide bonds in the cysteine-rich extracellular domain

    • Transient transfection or stable cell line generation using pCDNA3.1 or similar vectors with appropriate tags (His, Fc, or FLAG)

  • Insect Cell Expression Systems:

    • Baculovirus-infected Sf9 or High Five cells represent an alternative for higher yield

    • Suitable for the extracellular domain with proper post-translational modifications

  • E. coli Expression (Limited application):

    • Only recommended for specific protein fragments that do not require glycosylation

    • May be useful for cytoplasmic domain expression for structural studies

    • Requires refolding protocols when expressing disulfide-containing domains

Construct Design Considerations:

  • Express the extracellular domain (amino acids 1-184) for receptor-ligand interaction studies

  • Include the signal peptide sequence for proper processing in mammalian systems

  • Consider a C-terminal tag rather than N-terminal to avoid interference with signal peptide processing

  • For structural studies, remove flexible regions that might hinder crystallization

Purification Strategy:

  • Affinity Chromatography:

    • Immobilized metal affinity chromatography (IMAC) for His-tagged proteins

    • Protein A/G chromatography for Fc-fusion proteins

    • Anti-FLAG affinity chromatography for FLAG-tagged constructs

  • Further Purification Steps:

    • Ion exchange chromatography based on TNFRSF11A's isoelectric point

    • Size exclusion chromatography to remove aggregates and ensure monodispersity

    • Consider including low concentrations of reducing agents to prevent non-specific disulfide formation

  • Quality Control:

    • SDS-PAGE and Western blot to confirm identity and purity

    • Mass spectrometry to verify molecular weight and post-translational modifications

    • Circular dichroism to assess secondary structure

    • Thermal shift assays to evaluate protein stability

Functional Validation:

  • RANKL binding assays using surface plasmon resonance or bio-layer interferometry

  • Cell-based assays measuring NF-κB activation in response to the purified protein

  • Osteoclast differentiation assays to confirm biological activity

These methodological considerations should be adapted based on the specific research questions and downstream applications of the recombinant TNFRSF11A protein.

How can TNFRSF11A polymorphisms be effectively analyzed for association with bone disorders?

Analysis of TNFRSF11A polymorphisms and their association with bone disorders requires a systematic approach combining genetic, statistical, and functional methods. The following comprehensive methodology is recommended:

1. Polymorphism Identification and Genotyping:

  • Next-Generation Sequencing (NGS): Targeted sequencing of the TNFRSF11A gene, including exons, intron-exon boundaries, and regulatory regions

  • PCR-RFLP (Restriction Fragment Length Polymorphism): For known polymorphisms like IVS6+79A/G

  • TaqMan SNP Genotyping Assays: For high-throughput analysis of specific SNPs

  • Sanger Sequencing: For confirmation of novel variants

  • PAGE (Polyacrylamide Gel Electrophoresis): Particularly useful for detecting insertions/duplications that alter fragment size, as demonstrated in the FEO studies

2. Study Design Considerations:

  • Case-Control Studies: Compare genotype frequencies between patients with bone disorders and healthy controls

  • Family-Based Association Studies: Analyze inheritance patterns within families, as demonstrated in the FEO and PDB families

  • Meta-Analysis: Combine results from multiple studies to increase statistical power

  • Sample Size Calculation: Ensure adequate power to detect associations based on expected effect sizes

3. Statistical Analysis Approaches:

  • Hardy-Weinberg Equilibrium Testing: To check for genotyping errors or selection bias

  • Chi-Square or Fisher's Exact Test: For comparing genotype and allele frequencies between cases and controls

  • Logistic Regression: To calculate odds ratios and adjust for potential confounding factors

  • Haplotype Analysis: To evaluate combinations of polymorphisms that may act together

  • Linkage Disequilibrium Mapping: To identify regions of TNFRSF11A associated with disease

4. Functional Validation of Associated Polymorphisms:

  • Luciferase Reporter Assays: To assess the impact of promoter variants on gene expression

  • EMSA (Electrophoretic Mobility Shift Assay): To evaluate effects on transcription factor binding

  • RNA Stability Assays: For variants in untranslated regions

  • Cell-Based Functional Assays: To determine effects on NF-κB signaling, as performed for the FEO mutations

  • Animal Models: Generation of knock-in mice carrying human polymorphisms for in vivo functional studies

5. Clinical Correlation:

  • Genotype-Phenotype Correlation Analysis: Relating specific polymorphisms to clinical features

  • Bone Mineral Density Measurements: DXA scans to correlate genotypes with quantitative traits

  • Biomarker Analysis: Measuring bone turnover markers in relation to genotypes

  • Imaging Studies: Advanced imaging to characterize bone structure in different genotype groups

Example Application: IVS6+79A/G Polymorphism Analysis

This polymorphism was analyzed in 67 Northern-Irish patients with sporadic Paget's disease of bone and 63 age-matched controls . While the study did not find a significant association, the methodology demonstrates the principle of case-control design for TNFRSF11A polymorphism analysis.

By integrating these approaches, researchers can comprehensively evaluate the contribution of TNFRSF11A genetic variation to bone disorders, potentially identifying risk factors and novel therapeutic targets.

What experimental models are most appropriate for studying TNFRSF11A function in vivo?

Selecting appropriate experimental models is crucial for investigating TNFRSF11A function in vivo. Each model system offers distinct advantages and limitations for studying different aspects of RANK biology. The following models are recommended based on research objectives:

1. Mouse Models:

  • Global Knockout Models:

    • TNFRSF11A-/- mice display severe osteopetrosis due to the absence of osteoclasts, along with defects in lymph node development and mammary gland formation

    • These models are valuable for understanding the fundamental physiological roles of RANK but may have limitations for studying acquired bone disorders due to developmental defects

  • Conditional Knockout Models:

    • Cell-specific deletion using Cre-loxP system (e.g., cathepsin K-Cre for osteoclast-specific deletion)

    • Temporal control using inducible systems (e.g., tamoxifen-inducible CreERT2)

    • These models allow for the study of TNFRSF11A function in specific cell types and developmental stages, avoiding the confounding effects of developmental abnormalities

  • Knock-in Models:

    • Mice carrying human disease mutations such as the 84dup18 (FEO) or 75dup27 (PDB) duplications

    • These models recapitulate human disease pathogenesis and are valuable for testing therapeutic interventions

  • Transgenic Overexpression Models:

    • Overexpression of wild-type or constitutively active TNFRSF11A variants

    • Useful for studying gain-of-function effects and potential roles in cancer models

2. Rat Models:

  • Larger size facilitates certain procedures such as bone surgeries and imaging

  • Similar bone physiology to humans

  • Valuable for pharmacological studies due to more predictable drug metabolism

3. Zebrafish Models:

  • Rapid development and transparent embryos allow for real-time visualization of skeletal development

  • Amenable to high-throughput screening of chemical compounds targeting RANK signaling

  • CRISPR-Cas9 gene editing can be used to generate loss-of-function or knock-in mutations

4. Ex Vivo Bone Organ Cultures:

  • Calvarial organ cultures from neonatal mice

  • Maintain the complex cellular interactions within bone tissue

  • Suitable for short-term studies of RANK-mediated osteoclastogenesis and bone resorption

5. Humanized Mouse Models:

  • Immunodeficient mice reconstituted with human immune cells

  • Valuable for studying human-specific aspects of RANK function in immune regulation

  • Potential for testing human-specific therapeutics

Experimental Applications:

  • Bone Phenotyping:

    • Micro-CT analysis for bone microarchitecture

    • Histomorphometry for cellular parameters (osteoclast number, surface, etc.)

    • Biomechanical testing for functional consequences

  • Immune Function Assessment:

    • Flow cytometry analysis of dendritic cell and T cell populations

    • Lymph node development and organization

    • Immune responses to various challenges

  • Disease Modeling:

    • Inflammatory bone loss (e.g., arthritis models)

    • Metastatic bone disease models

    • Age-related bone loss

  • Therapeutic Testing:

    • Evaluation of RANK-targeted biologics

    • Assessment of small molecule inhibitors of downstream pathways

    • Testing of gene therapy approaches

Selection of the appropriate model should be guided by the specific research question, focusing on models that best recapitulate the human condition being studied while considering ethical principles and the 3Rs (Replacement, Reduction, Refinement) in animal research.

How do TNFRSF11A mutations contribute to the pathogenesis of bone disorders, and what are the clinical implications?

TNFRSF11A mutations play a central role in the pathogenesis of several bone disorders through dysregulation of osteoclast formation and function. Understanding these mechanisms has significant clinical implications for diagnosis, prognosis, and treatment.

Pathogenic Mechanisms:

  • Gain-of-Function Mutations:

    • Tandem duplications in the signal peptide region (84dup18 in FEO and 75dup27 in PDB) lead to constitutive activation of RANK-mediated NF-κB signaling

    • These mutations prevent normal cleavage of the signal peptide, resulting in altered protein processing and expression levels

    • Hyperactive RANK signaling promotes excessive osteoclast formation and activity, leading to enhanced bone resorption

    • The 84dup18 mutation has been identified in affected individuals from Northern Irish, American, and German FEO families, suggesting a common pathogenic mechanism

  • Loss-of-Function Mutations:

    • Associated with autosomal recessive osteopetrosis type 7

    • Result in impaired osteoclast formation and function, leading to increased bone density

    • May cause secondary hematopoietic and neurological complications due to reduced bone marrow space and nerve compression

Clinical Manifestations and Diagnostic Considerations:

  • Familial Expansile Osteolysis (FEO):

    • Progressive osteolytic lesions, particularly affecting the long bones

    • Early-onset deafness and tooth loss

    • Genetic testing for the 84dup18 mutation in TNFRSF11A is diagnostic

    • Differentiation from other osteolytic conditions including Paget's disease of bone

  • Paget's Disease of Bone (PDB):

    • Focal areas of increased bone turnover

    • Pain, deformity, and increased risk of fractures

    • TNFRSF11A mutations (e.g., 75dup27) identified in some familial cases

    • Multiple genetic loci implicated, with TNFRSF11A being one of several susceptibility genes

  • Osteopetrosis:

    • Increased bone density

    • Fractures despite increased bone mass due to poor bone quality

    • Hematopoietic failure

    • Neurological complications including vision and hearing loss

Therapeutic Implications:

  • Targeted Therapies:

    • Antiresorptive agents (bisphosphonates, denosumab) may be effective for conditions with increased osteoclast activity (FEO, PDB)

    • Recombinant RANKL might theoretically benefit patients with loss-of-function mutations

    • Development of small molecules targeting specific mutant RANK proteins or their downstream pathways

  • Personalized Medicine Approach:

    • Genetic testing to identify specific TNFRSF11A mutations

    • Tailoring treatment based on the molecular mechanism (gain vs. loss of function)

    • Monitoring response to therapy based on mutation status

  • Emerging Therapies:

    • Gene therapy approaches to correct specific mutations

    • Antisense oligonucleotides to modulate aberrant splicing

    • CRISPR-Cas9 gene editing for permanent correction of germline mutations

  • Genetic Counseling:

    • Identification of carriers within affected families

    • Preimplantation genetic diagnosis as an option for families with severe forms

    • Risk assessment for family members of affected individuals

Research in this area continues to evolve, with ongoing efforts to better understand genotype-phenotype correlations and develop more targeted therapeutic approaches based on the specific molecular defects in TNFRSF11A-associated bone disorders.

What is the evidence for TNFRSF11A involvement in cancer pathogenesis and potential as a therapeutic target?

TNFRSF11A (RANK) has emerged as a significant factor in cancer pathogenesis, with evidence supporting its potential as a therapeutic target in various malignancies. The following summarizes current research findings and translational implications:

Evidence for TNFRSF11A in Cancer Pathogenesis:

  • Expression in Cancer Cells:

    • TNFRSF11A is expressed in various cancer cell lines, including breast cancer cell lines such as MDA-MB-231, SKBR3, MCF7, MDA-MB-468, and T47D

    • Alternative splice variants of TNFRSF11A (RANK-a, RANK-b, RANK-c) show differential expression patterns across cancer cell lines

    • TNFRSF11A expression patterns correlate with clinical parameters such as tumor grade and proliferation index (Ki-67) in breast cancer samples

  • Functional Role in Tumor Cells:

    • RANK signaling promotes tumor cell survival and proliferation through activation of NF-κB and other pro-survival pathways

    • RANK-RANKL axis contributes to cancer cell migration and invasion

    • RANK signaling may induce epithelial-to-mesenchymal transition (EMT), enhancing metastatic potential

  • Role in Metastasis:

    • RANK-expressing tumor cells preferentially metastasize to bone, where RANKL is abundantly expressed

    • RANK-RANKL interaction in bone microenvironment promotes a "vicious cycle" of bone destruction and tumor growth

    • RANK signaling contributes to the formation of premetastatic niches

  • Impact of Splice Variants:

    • Studies demonstrate that TNFRSF11A_delta 7,8,9 (RANK-c) expression varies across different breast cancer cell lines, suggesting functional significance in cancer biology

    • Differential expression of RANK splice variants may contribute to tumor heterogeneity and treatment response

Methodological Approaches to Study TNFRSF11A in Cancer:

  • Expression Analysis:

    • RT-PCR and qPCR to quantify TNFRSF11A and splice variant expression in cancer cell lines and patient samples

    • Immunohistochemistry to evaluate RANK protein expression in tumor tissues

    • Western blotting to assess RANK protein levels and activation status of downstream signaling pathways

  • Functional Studies:

    • siRNA or CRISPR-Cas9 knockout to assess effects of RANK depletion on cancer cell behavior

    • Overexpression of wild-type RANK or specific splice variants to evaluate their role in cancer progression

    • In vitro assays for proliferation, migration, invasion, and resistance to apoptosis

  • In Vivo Models:

    • Orthotopic xenograft models to study the role of RANK in primary tumor growth

    • Metastasis models to evaluate its function in dissemination to distant sites, particularly bone

    • Genetic mouse models combining RANK alterations with cancer-driving mutations

Therapeutic Targeting Strategies:

  • Direct RANK Inhibition:

    • Anti-RANK monoclonal antibodies to block RANKL binding and subsequent signaling

    • Small molecule inhibitors of RANK dimerization or intracellular domain function

    • Peptide mimetics that disrupt RANK-RANKL interaction

  • Modulation of Splice Variants:

    • Antisense oligonucleotides to modify TNFRSF11A splicing patterns

    • Splice-switching oligonucleotides to promote expression of less oncogenic variants

    • Targeted degradation of cancer-associated splice variants

  • Combination Therapies:

    • RANK inhibitors with conventional chemotherapy

    • RANK-targeted agents with immune checkpoint inhibitors

    • Dual targeting of RANK and downstream effectors (e.g., NF-κB inhibitors)

  • Bone Metastasis Prevention:

    • Prophylactic use of RANK-targeted therapies in high-risk patients

    • Adjuvant therapy following primary tumor resection to prevent bone colonization

The emerging evidence for TNFRSF11A's involvement in cancer pathogenesis, particularly through alternative splicing mechanisms, represents a promising area for further research and therapeutic development. Clinical trials evaluating RANK-targeted approaches, either alone or in combination with established therapies, will be essential to translate these findings into improved patient outcomes.

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.