CXCL12 Antibody,HRP conjugated

Shipped with Ice Packs
In Stock

Description

Cancer Biology

  • Prostate Cancer: CXCL12/HRP antibodies identified elevated CXCL12 and CXCR4 expression in prostate tumors compared to benign tissue, correlating with perineural invasion and matrix metalloproteinase (MMP-2/MMP-9) activity .

  • Breast Cancer: Demonstrated CXCL12’s dual role in suppressing metastasis via CD8+ T-cell activation while promoting primary tumor growth inhibition .

  • Pancreatic/Colorectal Cancer: Detected covalent CXCL12–keratin-19 heterodimers coating cancer cells, influencing tumor microenvironment interactions .

Immunomodulation Studies

  • Dendritic Cell Function: CXCL12-treated monocytes showed impaired dendritic cell (DC) maturation and reduced antigen-specific T-cell activation .

  • Macrophage Differentiation: Antibodies validated CXCL12’s role in maintaining CD14+/CD4+ macrophage populations via RUNX3 suppression .

Sensitivity and Specificity

  • Western Blot: Detects CXCL12 at 1:500–1:2000 dilution, identifying isoforms (~10–15 kDa) .

  • Blocking Controls: Recombinant CXCL12 abolished staining in pancreatic cancer models, confirming antibody specificity .

Comparative Performance

FeatureCXCL12/HRP (OABF01499-HRP) Alternative Clone (ABIN6139271)
HostRabbit polyclonalRabbit polyclonal
EpitopeFull-length CXCL12AA 22–89
Cross-ReactivityHuman, Mouse, RatHuman, Mouse, Rat, Dog, Chicken
ApplicationsIHC-P, IFWB, IHC, IF

Critical Considerations

  • Sodium Azide Toxicity: Requires handling by trained personnel due to preservative content .

  • Batch Variability: Polyclonal nature may cause inter-lot reactivity differences; validation with recombinant CXCL12 is recommended .

  • Signal Optimization: Titration essential to balance background noise in IHC/IF .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery timelines may vary depending on the order fulfillment method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
12-O-tetradecanoylphorbol 13-acetate repressed protein 1 antibody; AI174028 antibody; C-X-C motif chemokine 12 antibody; Chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1) antibody; Chemokine (C-X-C motif) ligand 12 antibody; Chemokine CXC motif ligand 12 antibody; cxcl12 antibody; hIRH antibody; hSDF-1 antibody; Intercrine reduced in hepatomas antibody; IRH antibody; OTTHUMP00000019491 antibody; PBSF antibody; Pre-B cell growth-stimulating factor antibody; SCYB12 antibody; SDF 1 antibody; SDF-1 antibody; SDF-1-alpha(3-67) antibody; SDF-1a antibody; SDF-1b antibody; SDF1_HUMAN antibody; SDF1A antibody; SDF1B antibody; Stromal cell-derived factor 1 antibody; Stromal cell-derived factor 1 delta antibody; Stromal cell-derived factor 1 gamma antibody; Stromal cell-derived factor 1a antibody; Stromal cell-derived factor-1 alpha antibody; Thymic lymphoma cell-stimulating factor antibody; Tlsf antibody; TLSF-a antibody; TLSF-b antibody; Tlsfa antibody; Tlsfb antibody; TPAR1 antibody
Target Names
Uniprot No.

Target Background

Function

This CXCL12 antibody (HRP conjugated) is a chemoattractant acting on T-lymphocytes and monocytes, but not neutrophils. It activates the C-X-C chemokine receptor CXCR4, inducing a rapid, transient increase in intracellular calcium ion levels and subsequent chemotaxis. SDF-1β(3-72) and SDF-1α(3-67) exhibit reduced chemotactic activity. Binding to cell surface proteoglycans appears to inhibit SDF-1α(3-67) formation, thereby preserving activity at localized sites. This antibody also binds to the atypical chemokine receptor ACKR3, activating the β-arrestin pathway and functioning as a scavenger receptor for SDF-1. Furthermore, it binds to the allosteric site (site 2) of integrins, activating integrins ITGAV:ITGB3, ITGA4:ITGB1, and ITGA5:ITGB1 independently of CXCR4. It acts as a positive regulator of monocyte migration and a negative regulator of monocyte adhesion via LYN kinase. Monocyte and T-lymphocyte migration is stimulated through CXCR4 and ACKR3 receptors, while adherence to ICAM-1-coated surfaces (a β-2 integrin ligand) is decreased. The SDF1A/CXCR4 signaling axis inhibits β-2 integrin LFA-1-mediated monocyte adhesion to ICAM-1 through LYN kinase. This antibody also inhibits CXCR4-mediated infection by T-cell line-adapted HIV-1. It plays a protective role following myocardial infarction and induces the downregulation and internalization of ACKR3 expressed in various cells. CXCL12 has crucial roles in embryonic development, including B-cell lymphopoiesis, myelopoiesis in bone marrow, and heart ventricular septum formation. It also stimulates the proliferation of bone marrow-derived B-cell progenitors in the presence of IL7 and the growth of stromal cell-dependent pre-B-cells.

Gene References Into Functions

Relevant Research Findings: The following studies highlight the significant role of CXCL12 in various biological processes and diseases:

  1. Positive correlation between CXCL12 expression and reduced disease-free survival. PMID: 30182340
  2. CXCR7 and CXCR4's crucial roles in regulating hematopoietic cell migration and homing in response to CXCL12. PMID: 29433559
  3. Association between CXCL12 rs1801157 and human papillomavirus infection and high-grade intraepithelial lesions. PMID: 30227860
  4. Protective effect of CXCL12 silencing against podocyte injury, potentially through inhibition of the CXCL12/STAT3 signaling pathway. PMID: 29508174
  5. CXCL12/SDF1 protein expression as a favorable prognostic biomarker in breast cancer. PMID: 29800557
  6. The CXCL12-CXCR4 axis promotes migration, invasion, and EMT in B-CPAP cells, partly through activation of the NF-κB signaling pathway. PMID: 29316404
  7. CXCL12 methylation's role in epigenetic gene regulation in papillary thyroid carcinoma (PTC). PMID: 28272462
  8. Non-oxidizable HMGB1's sustained induction of cardiac fibroblast migration by altering the CXCL12/CXCR4 axis, impacting cardiac remodeling post-infarction. PMID: 28716707
  9. Regulation of CXCL12 gene transcription by its 3'UTR sequence and implications for diseases with abnormal CXCL12α expression. PMID: 30266500
  10. Association between high SDF-1 expression and bladder cancer progression. PMID: 30015971
  11. Association between high CXCL12 expression and metastasis in colon cancer. PMID: 29305742
  12. miR-125b's role as a downstream mediator of CXCL12/CXCR4 axis activation. PMID: 28176874
  13. Increased frequency of CXCL12-related rs18011517 polymorphism in non-Hodgkin lymphoma patients, potentially linked to pathogenesis and outcome. PMID: 30197351
  14. The crucial role of the CXCL12/CXCR4 axis in maintaining homeostasis, particularly during hematopoiesis, and its potential as a therapeutic target in hematological tumors. PMID: 29288743
  15. CXCL12-dependent migration of BCP-ALL cells towards mesenchymal stromal cells (MSCs). PMID: 28619846
  16. Elevated serum CXCR4 and CXCL12 levels in septic neonates as valuable diagnostic markers for neonatal sepsis. PMID: 28562124
  17. CXCL12 and CXCR4 polymorphisms as potential risk factors and markers for hepatocellular carcinoma (HCC). PMID: 29741398
  18. Stromal cell-derived factor-1/C-X-C chemokine receptor type 4 axis's induction of human dental pulp stem cell migration through FAK/PI3K/Akt and GSK3β/β-catenin pathways. PMID: 28067275
  19. EGFR overexpression and mutations' impact on human lung adenocarcinoma cells through the CXCR4/CXCL12 signaling pathway. PMID: 30037369
  20. Association between serum CXCL12 and head and neck squamous cell carcinomas. PMID: 29693336
  21. Abnormal expression of fibrosis markers, ERα, and the SDF-1/CXCR4 axis in intrauterine adhesions endometrium. PMID: 29568895
  22. Increased risk of developing late AIDS in HIV-1 infected individuals with SDF-1 3'A polymorphism. PMID: 30053458
  23. Involvement of the SDF1/CXCR4 signaling pathway in low-intensity pulsed ultrasound-promoted periodontal ligament stem cell migration. PMID: 29620151
  24. Allosteric integrin activation by SDF1 as a novel drug discovery target. PMID: 29301984
  25. Association between CXCL12 single nucleotide polymorphisms and hypertension risk in the Chinese Han population. PMID: 30180964
  26. Prognostic importance of the CXCR4-CXCL12 chemokine axis for breast cancer survival. PMID: 29516917
  27. Elevated serum SDF-1 as a potential marker for primary biliary cholangitis. PMID: 29414663
  28. CXCR4 antagonist AMD3100's disruption of the CXCR4/CXCL12 axis and its effect on liver metastasis. PMID: 29436696
  29. SDF-1α overexpression in bone marrow-derived stromal stem cells and its promotion of bone generation. PMID: 29758548
  30. SDF-1's induction of MMP expression in human endplate chondrocytes and its role in ECM remodeling in degenerated cartilage endplate. PMID: 29207021
  31. The interplay between VEGF-A and SDF-1 in EPC-mediated angiogenesis. PMID: 27943613
  32. Estrogen's promotion of ER-negative breast cancer progression by stimulating SDF-1α secretion from cancer-associated fibroblasts. PMID: 27996037
  33. Importance of SDF-1 and CXCR4 expression for loco-regional control and overall survival in HNSCC after primary radiochemotherapy. PMID: 29061496
  34. Adipocyte autocrine function involving SDF1 and its regulation of insulin resistance. PMID: 29581126
  35. SDF-1α's stimulation of E-selectin/ligand pair expression in endothelial cells and bone marrow-derived endothelial progenitor cells, enhancing EPC-EC interactions. PMID: 27713493
  36. CXCL12(5-67)'s lack of promotion of NSCs migration but induction of cell death. PMID: 28623786
  37. SDF-1/CXCR4-RhoA and RhoC-ROS-cytoskeleton pathway's regulation of Jurkat cell migration. PMID: 28536953
  38. Validation of mir31 expression upregulation using GEO data sets. PMID: 27597234
  39. Differential expression of SDF-1 receptor CXCR4 in inherited thrombocytopenias. PMID: 28032520
  40. The role of CXCL12 in bladder cancer. PMID: 29022185
  41. rhSDF-1α's acceleration of reendothelialization in aneurysm necks post flow diverter implantation. PMID: 28159982
  42. Increased invasiveness of cells at MELF pattern sites through CXCL14-CXCR4 and CXCL12-CXCR4 axes. PMID: 28277316
  43. SDF-1 rs1801157 polymorphism as a risk factor for cancer development, particularly urologic and lung cancers, among Asians. PMID: 27265091
  44. Protective effect of SDF1 against AIDS progression and death in some populations. PMID: 29420545
  45. Dependence of EMT, increased migration, and survival in MCF-7 and H460 cells on iron/ROS metabolism and CXCR4/CXCL12 axis. PMID: 28774348
  46. Elevated serum levels in preeclamptic women. PMID: 28001450
  47. Defect of CXCL12 promoter histone acetylation in colon cancer. PMID: 28418886
  48. Association between SDF-1α/CXCR4 signaling pathway and clinicopathological features and prognosis of nasopharyngeal carcinoma. PMID: 28559386
  49. CXCL12-CXCR7 axis's acceleration of pancreatic cancer cell migration and invasion through mTOR and Rho/ROCK pathways, and its association with poor prognosis. PMID: 27542220
  50. The role of CXCL12 in multiple sclerosis, focusing on serum concentrations and gene polymorphism at position +801. PMID: 27894110
Database Links

HGNC: 10672

OMIM: 600835

KEGG: hsa:6387

STRING: 9606.ENSP00000379140

UniGene: Hs.522891

Protein Families
Intercrine alpha (chemokine CxC) family
Subcellular Location
Secreted.
Tissue Specificity
Isoform Alpha and isoform Beta are ubiquitously expressed, with highest levels detected in liver, pancreas and spleen. Isoform Gamma is mainly expressed in heart, with weak expression detected in several other tissues. Isoform Delta, isoform Epsilon and i

Q&A

What is CXCL12 and why is it important in biological research?

CXCL12, also known as stromal cell-derived factor 1 (SDF-1), functions as a ligand for the G-protein coupled receptor CXCR4 and plays crucial roles in numerous biological processes. The protein participates in embryogenesis, immune surveillance, inflammatory responses, tissue homeostasis, and significantly influences tumor growth and metastasis . In immunological research, CXCL12 has gained prominence due to its involvement in directing cellular migration, particularly of immune cells like B-lymphocytes within germinal centers. The chemokine exists in multiple isoforms, with CXCL12α and CXCL12β being the most predominantly expressed variants, each potentially exhibiting distinct biological functions . Understanding CXCL12 signaling pathways provides insights into fundamental immune processes and pathological conditions including cancer metastasis and inflammatory disorders.

What detection methods are available for CXCL12 using antibodies?

CXCL12 can be detected through multiple immunological techniques, with antibody selection being crucial for experimental success. Immunohistochemistry (IHC) has been effectively used to detect CXCL12 in paraffin-embedded tissue sections, requiring appropriate antigen retrieval methods such as heat-mediated epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 8.0) . Immunofluorescence (IF) represents another powerful approach, typically employing fluorophore-conjugated secondary antibodies like Cy3-conjugated anti-rabbit IgG to visualize CXCL12 distribution in tissue sections. Western blotting can distinguish between CXCL12 isoforms, with specific antibodies demonstrating differential reactivity toward CXCL12α and CXCL12β . HRP-conjugated antibodies offer particular advantages for Western blotting and IHC applications due to their enhanced sensitivity and signal amplification capabilities when used with appropriate substrates.

How should CXCL12 expression be validated at the mRNA level?

Quantitative real-time PCR (qPCR) represents the gold standard for validating CXCL12 expression at the transcriptional level prior to protein analysis. Researchers should employ validated primer sets targeting conserved regions across CXCL12 isoforms, with SYBR Green-based detection methods providing reliable quantification . For optimal results, reactions should be performed in triplicates using cDNA generated from approximately 10 ng of RNA per reaction. The ΔΔCq method is recommended for quantification of CXCL12 mRNA expression relative to reference genes such as GAPDH. Establishing baseline expression across multiple cell lines can provide valuable controls for subsequent experiments, as demonstrated by studies identifying differential CXCL12 expression patterns in cell lines such as Caco-2 (high expression), HT-29 (no detectable expression), and A549 (very low expression) . This transcriptional profiling facilitates the identification of appropriate positive and negative control samples for antibody validation.

How should researchers validate the specificity of anti-CXCL12 antibodies?

Rigorous antibody validation is essential for obtaining reliable experimental data when working with CXCL12. Western blot analysis using recombinant CXCL12 isoforms represents a critical first step in validation, enabling assessment of antibody reactivity toward specific protein variants. Published comparisons of antibodies have revealed significant variability in specificity profiles; for instance, the rabbit polyclonal antibody LS-B943 reacts with CXCL12β but lacks broader specificity, while monoclonal antibody D8G6H demonstrates strong reactivity with CXCL12β and weaker detection of CXCL12α . Positive and negative cellular controls should be established through qPCR analysis of CXCL12 mRNA expression across multiple cell lines to identify appropriate cellular models. Cross-reactivity testing with related chemokines can further confirm specificity, particularly important when studying tissues expressing multiple chemokine family members. Researchers should also perform knockout/knockdown validation experiments when possible to definitively establish antibody specificity in complex biological samples.

What are the optimal protocols for CXCL12 detection in tissue samples?

Detection of CXCL12 in tissue samples requires careful optimization of immunohistochemical protocols. For paraffin-embedded sections, heat-mediated antigen retrieval is essential, with EDTA buffer (pH 8.0) demonstrating efficacy for human hepatitis tissue and citrate buffer (pH 6.0) working effectively for rat spleen tissues . Blocking with 10% goat serum minimizes non-specific binding, with primary antibody concentrations ranging from 1-5 μg/mL depending on the specific antibody and tissue type. For immunofluorescence applications, overnight incubation of primary antibody at 4°C followed by incubation with fluorophore-conjugated secondary antibodies (e.g., Cy3-conjugated goat anti-rabbit IgG) provides optimal signal-to-noise ratios. For chromogenic detection using HRP-conjugated antibodies, the Strepavidin-Biotin-Complex (SABC) system with DAB as the chromogen has demonstrated reliable results in detecting CXCL12 in spleen tissues . Counter-staining with DAPI or hematoxylin provides important contextual information regarding cellular and tissue architecture.

What factors influence the detection of different CXCL12 isoforms?

Detection of specific CXCL12 isoforms presents significant challenges requiring careful antibody selection and protocol optimization. Research indicates differential reactivity of antibodies toward CXCL12 isoforms, with most antibodies demonstrating stronger affinity for CXCL12β compared to CXCL12α, as demonstrated by Western blot analyses of recombinant proteins . This differential reactivity likely stems from structural variations between isoforms, particularly in the C-terminal regions. When analyzing tissue samples, researchers should consider that different tissues may preferentially express specific isoforms, necessitating selection of antibodies with appropriate isoform recognition profiles. Post-translational modifications including glycosylation and proteolytic processing can further complicate detection, potentially masking epitopes recognized by certain antibodies. Additionally, binding of CXCL12 to extracellular matrix components, particularly heparan sulfate proteoglycans, may alter antibody accessibility to epitopes in tissue samples . To comprehensively characterize CXCL12 expression, researchers may need to employ multiple antibodies recognizing distinct epitopes across different isoforms.

How can researchers effectively study CXCL12-mediated immune responses in cancer models?

Investigating CXCL12-mediated immune responses in cancer models requires multilayered experimental approaches examining both cellular and molecular parameters. Flow cytometric analysis of tumor-infiltrating lymphocytes and secondary lymphoid organs can reveal alterations in immune cell populations, with particular attention to CD8+ T cells, dendritic cells, and myeloid-derived suppressor cells, which have demonstrated significant responsiveness to CXCL12 manipulation . Functional assays including in vitro cytotoxicity tests comparing effector cells from control and CXCL12-modified tumor-bearing mice provide crucial insights into the cytolytic capacity of anti-tumor immune populations. Genetic approaches utilizing knockout models (e.g., perforin knockout or TRAIL knockout mice) can elucidate the mechanistic pathways through which CXCL12 influences anti-tumor immunity . Cytokine profiling, particularly measurement of IFN-γ, complements these approaches by identifying key soluble mediators of CXCL12-induced immune responses. Additionally, researchers should consider comparing results between immunocompetent and immunodeficient models to distinguish direct effects on tumor cells from immune-mediated activities, as exemplified by studies showing discrepant results between models with intact versus compromised immune systems.

What are the technical considerations for using CXCL12 antibodies in Western blotting for cancer research?

Western blotting with CXCL12 antibodies in cancer research requires specific technical optimizations to ensure reliable detection of this low molecular weight chemokine. Sample preparation is critical, with protein extraction methods needing to effectively recover secreted proteins that may be bound to extracellular matrix components. Depending on experimental questions, researchers may need to analyze both cellular lysates and conditioned media to capture both intracellular and secreted CXCL12. Due to the small size of CXCL12 (approximately 8-12 kDa depending on the isoform), high percentage (15-18%) polyacrylamide gels are recommended for optimal resolution of protein bands . When using HRP-conjugated antibodies, enhanced chemiluminescence detection systems provide the sensitivity needed for detecting physiological CXCL12 expression levels. Importantly, validation studies have shown differential reactivity of antibodies toward specific CXCL12 isoforms, with monoclonal antibody D8G6H demonstrating stronger reactivity to CXCL12β compared to CXCL12α . This differential recognition necessitates careful antibody selection based on the specific isoforms expressed in the cancer model under investigation. Positive controls using recombinant CXCL12 isoforms should be included to confirm antibody performance in each experimental run.

How does immobilized CXCL12 regulate germinal center reactions and antibody production?

Immobilized CXCL12 plays a fundamental role in regulating B-lymphocyte positioning during germinal center (GC) reactions, a process critical for generating high-affinity antibodies. Research using CXCL12gagtm mice, which express CXCL12 that cannot bind to cellular or extracellular surfaces, has demonstrated that immobilized CXCL12 establishes a fixed gradient with higher concentrations in the dark zone (DZ) of germinal centers . This gradient, in opposition to CXCL13 gradients, directs B-cell migration between the dark zone and light zone (LZ) through alternating expression of CXCR4. Disruption of CXCL12 binding to heparan sulfate prevents the establishment of this fixed gradient, impairing the mechanism of stepwise selection for B cells with increasing affinity to antigens. Consequently, CXCL12gagtm mice demonstrate poor DZ/LZ segregation within germinal centers and aberrant localization of mitotic cells, which are normally restricted to the dark zone compartment . These structural alterations ultimately result in reduced accumulation of somatic mutations in immunoglobulin genes and impaired affinity maturation, highlighting the essential role of immobilized CXCL12 in optimal humoral immune responses.

What methods can assess CXCL12-dependent B cell migration and germinal center organization?

Investigating CXCL12-dependent B cell migration and germinal center organization requires specialized techniques spanning from molecular to histological analyses. Immunohistochemical evaluation of lymphoid follicles using markers for germinal center compartmentalization (e.g., Ki67 for proliferating centroblasts in the dark zone) provides critical information about structural organization . Laser capture microdissection coupled with qPCR allows quantification of chemokine gradients across germinal center compartments. Somatic hypermutation analysis represents a powerful approach to assess the functional impact of CXCL12 alterations, requiring sequencing of immunoglobulin variable regions from sorted GC B cells to determine mutation frequency and patterns. For example, sequencing a 294 bp region of the VH186.2 gene from λ1+IgG1+ GC B cells following NP-CGG immunization revealed reduced mutation accumulation in CXCL12gagtm mice compared to controls (median 4 versus 5 mutations per sequence) . High-affinity mutation acquisition, such as the W33L substitution in the VH186.2 gene that confers 10-fold increased affinity for NP, can serve as a functional readout of proper germinal center dynamics. Additionally, serum antibody titers and affinity measurements complete the analysis by confirming the functional consequences of altered CXCL12 activity on humoral immunity.

What are common pitfalls in CXCL12 antibody-based experiments and how can they be addressed?

Researchers frequently encounter several challenges when working with CXCL12 antibodies that require specific troubleshooting approaches. Antibody specificity issues represent a primary concern, as studies have demonstrated variable recognition of CXCL12 isoforms among commercially available antibodies . For instance, while the monoclonal antibody D8G6H exhibits strong reactivity with CXCL12β, it shows weaker detection of CXCL12α, potentially leading to false negative results when analyzing tissues predominantly expressing the latter isoform. Background signal, particularly in immunohistochemical applications, can compromise data interpretation and may be addressed through optimized blocking procedures using 10% goat serum and careful titration of primary antibody concentrations . Detection sensitivity limitations, especially relevant for tissues with low CXCL12 expression, can be mitigated by employing signal amplification systems such as the Strepavidin-Biotin-Complex (SABC) for chromogenic detection or tyramide signal amplification for fluorescence-based methods. Batch-to-batch variability in antibody performance necessitates inclusion of standardized positive controls in each experimental run, ideally using validated cell lines with confirmed CXCL12 expression profiles like Caco-2 cells . Additionally, researchers should be aware that CXCL12 binding to extracellular matrix components may mask epitopes, potentially requiring optimization of antigen retrieval methods to expose the target protein fully.

How can researchers optimize CXCL12 detection in challenging tissue samples?

Detection of CXCL12 in challenging tissue samples requires systematic optimization of immunohistochemical protocols to overcome tissue-specific obstacles. For formalin-fixed paraffin-embedded tissues, extended antigen retrieval periods (20 minutes or longer) using either citrate buffer (pH 6.0) or EDTA buffer (pH 8.0) may be necessary to adequately expose CXCL12 epitopes . In tissues with high endogenous peroxidase activity, such as liver, additional quenching steps with hydrogen peroxide treatment can minimize background when using HRP-conjugated detection systems. Dual immunofluorescence approaches combining CXCL12 staining with markers of expected producer cells (e.g., stromal cells, endothelial cells) can increase confidence in signal specificity and provide contextual information about the source of CXCL12 within the tissue microenvironment. For tissues with known high protease activity, addition of protease inhibitors during sample processing helps preserve CXCL12 integrity. Comparison of multiple antibodies targeting different epitopes can overcome issues related to epitope masking or post-translational modifications. Additionally, for very low abundance detection, researchers might consider employing proximity ligation assays (PLA) which provide signal amplification for detecting CXCL12-receptor interactions with single-molecule sensitivity, offering insights into chemokine functionality beyond mere presence.

What controls should be included in CXCL12 antibody experiments to ensure validity?

Inclusion of appropriate controls is imperative for ensuring the validity of CXCL12 antibody experiments across all applications. Positive controls should include recombinant CXCL12 protein standards (preferably both CXCL12α and CXCL12β isoforms for Western blotting) and validated cell lines with confirmed CXCL12 expression, such as Caco-2, which exhibits high CXCL12 mRNA levels . Negative controls should incorporate cell lines with negligible CXCL12 expression (e.g., A549 or HT-29) and isotype-matched control antibodies to assess non-specific binding. For tissue immunohistochemistry, omission of primary antibody while maintaining all other steps provides crucial information about potential endogenous peroxidase activity or non-specific binding of detection reagents. Competition assays, where pre-incubation of the antibody with recombinant CXCL12 should eliminate specific staining, offer powerful validation of signal specificity. When possible, CXCL12 knockout or knockdown samples represent gold standard negative controls, though these may not be readily available for all experimental systems. For functional studies investigating CXCL12-dependent processes, neutralizing antibodies against CXCR4 or small molecule CXCR4 antagonists (e.g., AMD3100) provide important controls to confirm receptor specificity . Additionally, for studies focusing on immobilized versus soluble CXCL12, comparison between wild-type and CXCL12gagtm models offers valuable insights into functional distinctions between these forms .

How are CXCL12 antibodies being integrated with advanced imaging technologies?

Integration of CXCL12 antibodies with cutting-edge imaging technologies is expanding our understanding of chemokine distribution and function in complex tissues. Super-resolution microscopy techniques including Stimulated Emission Depletion (STED) and Stochastic Optical Reconstruction Microscopy (STORM) now allow visualization of CXCL12 gradients at nanoscale resolution, revealing previously unappreciated microanatomical details of chemokine immobilization patterns. Intravital multiphoton microscopy combined with fluorescently-tagged anti-CXCL12 antibodies enables real-time tracking of chemokine-guided cell migration in living tissues, particularly valuable for studying dynamic processes like lymphocyte trafficking in germinal centers . Mass cytometry imaging (e.g., Imaging Mass Cytometry or Multiplexed Ion Beam Imaging) permits simultaneous detection of CXCL12 alongside dozens of other proteins in single tissue sections, providing unprecedented insights into the relationship between chemokine expression and cellular niches within the tissue microenvironment. CLARITY and other tissue clearing methods combined with whole-mount immunostaining for CXCL12 allow three-dimensional reconstruction of chemokine gradients throughout intact organs. Additionally, correlative light and electron microscopy approaches make it possible to connect CXCL12 immunolabeling with ultrastructural features of producing and responding cells, offering unique perspectives on the subcellular organization of chemokine signaling components.

What novel applications are emerging for CXCL12 antibodies in translational research?

CXCL12 antibodies are finding novel applications in translational research spanning multiple disease contexts. In oncology, multiplex immunohistochemistry panels incorporating CXCL12 alongside immune cell markers are being developed to characterize tumor immune microenvironments and potentially predict response to immunotherapies. The observed ability of CXCL12 to inhibit tumor growth and metastasis through immune-mediated mechanisms in breast cancer models suggests potential therapeutic applications . Importantly, monitoring shifts in CXCL12 gradients during disease progression may provide prognostic information, as disruption of normal chemokine patterns can significantly impact immune cell positioning and function. In regenerative medicine, CXCL12 antibodies are helping elucidate the role of this chemokine in stem cell homing and tissue repair processes, potentially informing development of therapies that manipulate CXCL12 gradients to enhance healing. Autoimmune disease research is benefiting from insights into how CXCL12-directed cell positioning influences germinal center reactions and antibody affinity maturation, with implications for understanding diseases characterized by pathogenic autoantibody production . Additionally, CXCL12 detection in cerebrospinal fluid and brain tissue is providing insights into neuroinflammatory conditions, where alterations in this chemokine may contribute to aberrant immune cell trafficking within the central nervous system.

How might artificial intelligence and computational approaches enhance CXCL12 antibody research?

The integration of artificial intelligence and computational approaches with CXCL12 antibody research promises to transform our understanding of chemokine biology and experimental methodologies. Machine learning algorithms applied to immunohistochemical image analysis can quantify CXCL12 expression patterns across tissue samples with greater precision and reproducibility than manual scoring, potentially revealing subtle alterations associated with disease states. Deep learning approaches trained on large datasets of validated CXCL12 staining patterns could assist in antibody validation by automatically detecting non-specific binding or background issues. Computational modeling of CXCL12 gradients within tissues, informed by quantitative immunohistochemistry data, can simulate cell migration dynamics and predict outcomes of gradient perturbations, generating testable hypotheses about chemokine function. Network analysis algorithms incorporating CXCL12 signaling within larger molecular interaction maps help contextualize experimental findings within broader biological processes. AI-assisted epitope prediction tools can identify optimal target regions for developing novel CXCL12 antibodies with enhanced specificity for particular isoforms or post-translationally modified variants. Furthermore, natural language processing of the scientific literature could accelerate discovery by automatically extracting and synthesizing information about CXCL12 across thousands of publications, identifying connections and research gaps that might otherwise remain obscure. Together, these computational approaches hold potential to dramatically accelerate research progress and generate novel insights from CXCL12 antibody-based experiments.

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 2025 TheBiotek. All Rights Reserved.