CXCL12 Antibody

Shipped with Ice Packs
In Stock

Description

Mechanisms of Action

CXCL12 antibodies exert effects through:

  • Neutralization of CXCL12: Blocks binding to CXCR4/CXCR7, inhibiting downstream signaling .

  • Immune Modulation: Reduces CD8+ T-cell activation and dendritic cell-mediated antigen presentation .

  • Angiogenesis Suppression: Downregulates VEGF and CCL1, key proangiogenic factors .

In melanoma, CXCL12 antibodies disrupt autocrine signaling in tumor-associated macrophages (TAMs), which constitute 60–90% of TAMs in metastatic tissues . In alopecia areata (AA) models, humanized CXCL12 antibodies reduce CD8+ T-cell infiltration and suppress Jak/Stat pathway activation .

Cancer Therapeutics

  • Preclinical Efficacy: Humanized antibody hu30D8 inhibited primary tumor growth and metastasis in xenograft models .

  • Synergy with Anti-VEGF: Combined therapy enhanced antitumor effects in orthotopic models .

Autoimmune and Inflammatory Diseases

  • Collagen-Induced Arthritis: hu30D8 reduced disease severity by 50% in mice, with additive effects when combined with TNF-α inhibitors .

  • Alopecia Areata: Treatment delayed disease onset by reducing dendritic cells and CD8+ T-cells in skin lesions .

Reproductive Health

  • Endometrial Receptivity: Intrauterine CXCL12 administration improved embryo implantation rates in mice by enhancing integrin β3 and osteopontin expression .

Comparative Applications

ApplicationMechanismOutcomeReference
Metastatic MelanomaDisrupts TAM proangiogenic signalingReduced tumor-associated angiogenesis
Alopecia AreataSuppresses CD8+ T-cell activationDelayed hair loss onset
Choroidal NeovascularizationInhibits CXCL12-driven vascular growth50% reduction in lesion size

Challenges and Future Directions

  • Dosing Optimization: Varied clearance rates across species necessitate tailored regimens .

  • Combination Therapies: Synergy with checkpoint inhibitors or anti-angiogenics remains underexplored .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
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
CXCL12 is a chemoattractant that primarily targets T-lymphocytes and monocytes, but not neutrophils. It activates the CXCR4 receptor, triggering a rapid and transient increase in intracellular calcium ion levels and chemotaxis. Variants like SDF-1-beta(3-72) and SDF-1-alpha(3-67) exhibit reduced chemotactic activity. CXCL12's interaction with cell surface proteoglycans appears to inhibit the formation of SDF-1-alpha(3-67), thereby preserving its activity at local sites. It also binds to the atypical chemokine receptor ACKR3, which activates the beta-arrestin pathway and acts as a scavenger receptor for SDF-1. CXCL12 binds to the allosteric site (site 2) of integrins, activating integrins such as ITGAV:ITGB3, ITGA4:ITGB1, and ITGA5:ITGB1 independently of CXCR4. It serves as a positive regulator of monocyte migration and a negative regulator of monocyte adhesion through the LYN kinase pathway. CXCL12 promotes the migration of monocytes and T-lymphocytes via its receptors, CXCR4 and ACKR3, and concurrently reduces monocyte adhesion to surfaces coated with ICAM-1, a ligand for beta-2 integrins. The SDF1A/CXCR4 signaling axis inhibits the beta-2 integrin LFA-1 mediated adhesion of monocytes to ICAM-1 by engaging the LYN kinase. CXCL12 effectively inhibits CXCR4-mediated infection by T-cell line-adapted HIV-1. It plays a crucial protective role in the aftermath of myocardial infarction. CXCL12 induces downregulation and internalization of ACKR3 expressed in various cell types. It plays critical roles during embryonic development, being essential for B-cell lymphopoiesis, myelopoiesis in the bone marrow, and the formation of the heart ventricular septum. CXCL12 stimulates the proliferation of bone marrow-derived B-cell progenitors in the presence of IL7, as well as the growth of stromal cell-dependent pre-B-cells.
Gene References Into Functions
  1. Cases with positive CXCL12 expression exhibited shorter disease-free survival rates compared to cases with negative CXCL12 expression. PMID: 30182340
  2. This study highlights the crucial role of CXCR7, alongside CXCR4, in regulating normal and malignant hematopoietic cell migration and homing induced by CXCL12. PMID: 29433559
  3. CXCL12 rs1801157 is independently associated with Human papillomavirus infection and influences the development of high-grade intraepithelial lesions. PMID: 30227860
  4. Silencing CXCL12 demonstrates a protective effect against podocyte injury, potentially by inhibiting the CXCL12/STAT3 signaling pathway. PMID: 29508174
  5. The CXCL12/SDF1 protein expression serves as a valuable prognostic biomarker in breast cancer. PMID: 29800557
  6. The CXCL12-CXCR4 axis drives the migration, invasion, and epithelial-mesenchymal transition processes in B-CPAP cells, at least partially, through activation of the NF-kappaB signaling pathway. PMID: 29316404
  7. This study reveals CXCL12 methylation-mediated epigenetic regulation of gene expression in papillary thyroid carcinoma (PTC). This research was the first to perform reduced representation bisulfite sequencing analysis for PTC, suggesting that CXCL12 may contribute to PTC development through methylation-mediated epigenetic regulation of gene expression. PMID: 28272462
  8. These results show that non-oxidizable HMGB1 induces sustained cardiac fibroblasts migration, irrespective of the redox state of the environment, by modulating the CXCL12/CXCR4 axis. This impacts proper cardiac remodeling following infarction. PMID: 28716707
  9. This provides a foundation for understanding how multiple elements in the sequence encoding the 3'UTR of the CXCL12 gene regulate its transcription and offers insights into diseases involving abnormal CXCL12alpha expression. PMID: 30266500
  10. High SDF-1 expression is associated with the progression of bladder cancer. PMID: 30015971
  11. High CXCL12 expression is linked to metastasis in colon cancer. PMID: 29305742
  12. MiR-125b functions as a significant downstream mediator upon activation of the CXCL12/CXCR4 axis. PMID: 28176874
  13. The CXCL12-related rs18011517 polymorphism was more prevalent in non-Hodgkin lymphoma patients, suggesting a potential association with non-Hodgkin lymphoma pathogenesis and outcome. PMID: 30197351
  14. Data suggests that CXCL12 and its receptor CXCR4 play critical roles in maintaining homeostasis, particularly during hematopoiesis. Current clinical trials, especially in hematological tumors, are investigating whether incorporating CXCR4 inhibitors to hinder tumor dissemination will enhance the effectiveness of ongoing anti-cancer treatments. (CXCL12 = C-X-C motif chemokine ligand 12; CXCR4 = C-X-C motif chemokine receptor-4) [REVIEW] PMID: 29288743
  15. BCP-ALL cells actively migrate towards mesenchymal stromal cells (MSCs) in a CXCL12-dependent manner. PMID: 28619846
  16. Serum CXCR4 and CXCL12 levels increase significantly in septic neonates, making them valuable diagnostic markers for neonatal sepsis. Serum concentrations of both chemokines represent promising novel biomarkers for this condition. PMID: 28562124
  17. CXCL12 and CXCR4 polymorphisms may serve as risk factors for hepatocellular carcinoma (HCC), and they may be potential HCC markers. PMID: 29741398
  18. The stromal cell derived factor-1/C-X-C chemokine receptor type 4 axis induces human dental pulp stem cell migration through FAK/PI3K/Akt and GSK3beta/beta-catenin pathways. PMID: 28067275
  19. EGFR overexpression and mutations lead to alterations in the biological characteristics of human lung adenocarcinoma cells through the CXCR4/CXCL12 signaling pathway. PMID: 30037369
  20. Serum CXCL12, but not CXCR4, is associated with Head and Neck Squamous Cell Carcinomas. PMID: 29693336
  21. The aim of this study was to assess whether fibrosis markers, estrogen receptor (ER)alpha and the stromal derived factor (SDF)1/CXC chemokine receptor type 4 (CXCR4) axis are abnormally expressed in Intrauterine adhesions endometrium. PMID: 29568895
  22. HIV-1 infected individuals with the SDF-1 3'A polymorphism have a higher likelihood of developing late-stage AIDS. PMID: 30053458
  23. The SDF1/CXCR4 signaling pathway plays a role in Low-intensity pulsed ultrasound-promoted periodontal ligament stem cell migration. PMID: 29620151
  24. These findings suggest that SDF1, when presented on proteoglycans, can rapidly activate integrins in an allosteric manner by binding to site 2 in the absence of CXCR4. The allosteric integrin activation by SDF1 represents a novel target for drug discovery. PMID: 29301984
  25. An association between CXCL12 single nucleotide polymorphisms and the risk of hypertension in the Chinese Han population was observed. PMID: 30180964
  26. These results highlight the importance of the CXCR4-CXCL12 chemokine axis in breast cancer progression and emphasize the prognostic significance of this chemokine axis for breast cancer survival. PMID: 29516917
  27. Serum SDF-1 is elevated in primary biliary cholangitis and may serve as a potential useful marker for this condition. PMID: 29414663
  28. Disrupting the CXCR4/CXCL12 axis with the CXCR4 antagonist AMD3100 blocked the contributions of both cancer and stromal cells to the metastatic cascade in the liver. PMID: 29436696
  29. SDF-1 alpha overexpression in bone marrow-derived stromal stem cells promotes bone generation as indicated by enhanced osteogenesis and angiogenesis. PMID: 29758548
  30. These findings suggest that SDF1, as an inflammatory cytokine, induces MMP expression in human endplate chondrocytes, and that ECM remodeling in the degenerated cartilage endplate may be a favorable factor for endogenous stem cell homing into the nucleus pulposus for regeneration in vivo. PMID: 29207021
  31. Data demonstrate that: (i) hypoxia does not affect the capacity of EPCs to support the angiogenic process; (ii) the absence of either VEGF-A or SDF-1 from EPC-CM can be compensated by the presence of the other, so that the overall angiogenic effects remain unchanged; and (iii) the concomitant deletion of VEGF-A and SDF-1 from EPC-CM impairs its pro-angiogenic effect, both in vitro and in vivo. PMID: 27943613
  32. Estrogen may promote the progression of ER-negative breast cancer by stimulating cancer-associated fibroblasts to secrete SDF-1alpha, which can recruit MDSCs to the tumor microenvironment to exert tumor-promoting effects. PMID: 27996037
  33. Data supports the importance of SDF-1 and CXCR4 expression for loco-regional control and overall survival in HNSCC after primary radiochemotherapy. PMID: 29061496
  34. Data (including data from studies in knockout mice) suggest that adipocyte autocrine function involving SDF1 regulates insulin resistance; SDF1 gene expression correlates with insulin-desensitized conditions in adipocytes but not other tissues (liver, skeletal muscle); adipocyte-specific ablation of Sdf1 enhances insulin sensitivity in adipose tissues and in the whole body. PMID: 29581126
  35. This study reports that stromal cell-derived factor-1alpha elevated or therapeutically administered in ischemic wounded tissue can stimulate both local endothelial cells (EC) and bone marrow-derived endothelial progenitor cells (EPC) to reciprocally express E-selectin/ligand pairs, thereby enhancing EPC-EC interactions. PMID: 27713493
  36. Authors produced recombinant CXCL12 and CXCL12(5-67) and evaluated their effects on murine adult NSCs migration and survival in vitro. The study showed that CXCL12(5-67) does not promote NSCs migration but induces cell death. PMID: 28623786
  37. A SDF-1/CXCR4-RhoA and RhoC-ROS-cytoskeleton pathway regulating Jurkat cell migration in response to SDF-1 has been identified. PMID: 28536953
  38. Expression upregulation of mir31 was also validated using GEO data sets. PMID: 27597234
  39. Differential expression of the SDF-1 receptor CXCR4 in molecularly defined forms of inherited thrombocytopenias has been observed. PMID: 28032520
  40. A review of the role of CXCl12 in bladder cancer. PMID: 29022185
  41. Intravenous administration of rhSDF-1alpha accelerates reendothelialization in the aneurysm neck after flow diverter implantation. PMID: 28159982
  42. These findings suggest that cells at the sites of MELF pattern had acquired increased invasiveness through the function of the CXCL14-CXCR4 and CXCL12-CXCR4 axes. PMID: 28277316
  43. This study suggested that the SDF-1 rs1801157 polymorphism may serve as a risk factor for cancer development among Asians, particularly an increased risk of urologic and lung cancers. PMID: 27265091
  44. No significant association was found between SDF1 polymorphism and HIV susceptibility; a protective effect of SDF1 on AIDS progression and death was observed; in conclusion, SDF1 polymorphism exerts a moderate protective effect against AIDS disease deterioration in some specific populations. PMID: 29420545
  45. Findings indicate that induction of EMT, increased migration, and survival depend, in MCF-7 and H460 cells, on the release of FHC control on two pathways: namely the iron/ROS metabolism and CXCR4/CXCL12 axis. PMID: 28774348
  46. Serum levels are higher in preeclamptic women. PMID: 28001450
  47. A defect in CXCL12 promoter histone acetylation may represent an additional process involved in CXCL12 expression extinction in colon cancer. PMID: 28418886
  48. These findings indicated that the SDF-1alpha/CXCR4 signaling pathway might be associated with the clinicopathological features and prognosis of patients with nasopharyngeal carcinoma. PMID: 28559386
  49. The CXCL12-CXCR7 axis accelerates the migration and invasion of pancreatic cancer cells through the mTOR and Rho/ROCK pathways, and predicts poor prognosis for pancreatic cancer. PMID: 27542220
  50. A review of the role of CXCL12 in multiple sclerosis, with an emphasis on CXCL12 serum concentrations and its gene polymorphism at position +801. PMID: 27894110

Show More

Hide All

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

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.O
By Olaf Bartsch
★★★★★

Applications : Western Blot (WB)

Sample type: alpha, beta and gamnma SFD-1 subunits as purified CXCL12 proteins

Sample dilution: 1:1000

Review: The antibody does only detect alpha-subunits of CXCL12, but not beta and/or gamma- subunits. This was requested (customer was looking for an anti-CXCL12 alpha/beta/gamma) and customer performed such tests as promised. Attached please find the corresponding WB data.

Q&A

What is CXCL12 and what are its primary biological functions?

CXCL12 (C-X-C Motif Chemokine Ligand 12) functions as a ligand for the G-protein coupled receptor, chemokine (C-X-C motif) receptor 4 (CXCR4). This protein plays diverse roles across multiple biological processes including embryogenesis, immune surveillance, inflammatory responses, tissue homeostasis, and tumor growth and metastasis . Traditionally classified as a homeostatic chemokine, CXCL12 contributes to critical physiological processes such as embryogenesis, hematopoiesis, and angiogenesis . The CXCL12/CXCR4/ACKR3 axis constitutes a potential therapeutic target for numerous inflammatory diseases, not only by regulating cell migration but also by modulating immune responses . Understanding these fundamental functions provides the foundation for research applications of CXCL12 antibodies.

In which tissues is CXCL12 commonly expressed?

CXCL12 demonstrates widespread expression across multiple tissue types, making antibody validation in specific tissues an important consideration. According to expression profiles from Uniprot.org and published literature, CXCL12 is expressed in myometrium, fetal brain, fetal heart, heart, fibroblasts, liver, thymus, uterus, and brain, among other tissues . In the skin, CXCL12 is highly expressed in dermal fibroblasts (DFs) and has been implicated in mediating inflammatory skin diseases . The broad expression pattern explains why CXCL12 antibodies are applicable in diverse research contexts, from neurological studies to immunological investigations. This wide distribution necessitates careful consideration of tissue-specific validation when selecting a CXCL12 antibody for a particular experimental system.

What are the common applications for CXCL12 antibodies in research?

CXCL12 antibodies are commonly used in several experimental applications including Western blotting (WB), enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC) across human, mouse, and rat tissues . Beyond these standard applications, CXCL12-neutralizing antibodies have demonstrated efficacy in delaying disease onset or preventing disease progression in models of cancer, viral infections, inflammatory bowel diseases, rheumatoid arthritis, and osteoarthritis . In research settings, these antibodies serve both as analytical tools to detect CXCL12 expression and as therapeutic agents to modulate CXCL12-mediated processes. The versatility of CXCL12 antibodies makes them valuable tools for investigating both physiological and pathological processes across multiple experimental systems.

How should researchers validate CXCL12 antibody specificity for their tissue of interest?

Validating CXCL12 antibody specificity requires a systematic approach, particularly when applying the antibody to a new tissue type or species. Based on established practices, researchers should first consult literature and antibody databases to determine if the specific antibody has been validated in their tissue of interest. For example, CXCL12 antibody A00053-2 has been validated for ELISA, IHC, and WB in human, mouse, and rat tissues . When working with unvalidated tissue types, researchers should employ multiple techniques to confirm specificity: (1) Western blotting to verify the correct molecular weight, (2) positive controls using tissues known to express CXCL12 (e.g., liver or brain), (3) negative controls through antibody blocking with recombinant CXCL12 protein, and (4) correlation with mRNA expression data where possible. Additionally, researchers can leverage cross-validation with different antibody clones targeting distinct epitopes of CXCL12 to further confirm specificity.

What protocols are recommended for CXCL12 antibody usage in Western blotting?

For optimal Western blotting results with CXCL12 antibodies, researchers should follow validated protocols that address the specific characteristics of this chemokine. Based on established methodologies, CXCL12 typically requires proper sample preparation to maintain protein integrity. Researchers should homogenize tissues in RIPA buffer supplemented with protease inhibitors, considering CXCL12's relatively small size (8-14 kDa depending on isoform). Standard SDS-PAGE should utilize higher percentage gels (12-15%) to resolve this small protein effectively. For transfer, PVDF membranes are often preferred over nitrocellulose for their protein retention capabilities. After blocking (typically with 5% non-fat milk or BSA in TBST), researchers should incubate with primary CXCL12 antibody at manufacturer-recommended dilutions (typically 1:500-1:2000) overnight at 4°C. Detection systems should be selected based on the expected expression level, with chemiluminescence offering good sensitivity for most applications. Importantly, proper positive controls (recombinant CXCL12 or tissues with known expression) should be included to validate results .

How can researchers determine if a CXCL12 antibody recognizes specific isotypes or splice variants?

Determining CXCL12 antibody specificity for particular isotypes or splice variants requires detailed understanding of the antibody's immunogen and targeted epitope. CXCL12 exists in multiple splice variants (α, β, γ, δ, ε, and θ) with differing tissue distributions and functional properties. To assess isotype specificity, researchers should first identify the immunogen used for antibody generation. For example, the immunogen of antibody A00053-2 is E. coli-derived human CXCL12 recombinant protein (Position: V24-M93) . This information helps determine which domain of CXCL12 is recognized and potentially which isotypes would be detected. Experimental validation should include: (1) Western blotting with recombinant proteins representing different CXCL12 isotypes, (2) expression analysis in tissues known to preferentially express specific isotypes, and (3) blocking studies with isotype-specific peptides. Additionally, researchers can employ knockout/knockdown models or isotype-specific siRNAs followed by antibody testing to confirm specificity. When isotype specificity is critical to the research question, using multiple antibodies targeting different epitopes provides more comprehensive characterization.

How does CXCL12 antibody treatment affect immune cell populations and gene expression in inflammatory disease models?

CXCL12 antibody treatment significantly modulates immune cell populations and associated gene expression patterns in inflammatory disease models, as demonstrated in recent single-cell RNA sequencing (scRNA-seq) studies. In an alopecia areata (AA) mouse model, CXCL12 antibody treatment notably reduced the proportions of CD8+ T cells and dendritic cells/macrophages that were significantly increased in the disease state . At the transcriptional level, the antibody normalized approximately 78% of differentially expressed genes (DEGs) that were dysregulated in the AA model . Specifically, 153 DEGs that increased in the AA model subsequently decreased following antibody treatment, representing key mediators of both disease pathogenesis and therapeutic response .

Pathway analysis revealed that CXCL12 antibody treatment significantly downregulated processes upregulated in the disease state, including:

  • Immune cell chemotaxis (lymphocytes and monocytes)

  • Chemokine-mediated signaling

  • Cellular response to type II interferon

  • Regulation of leukocyte differentiation

Key immune-related genes modulated by CXCL12 antibody treatment included Ifng, Cd8a, Ccr5, Ccl4, Ccl5, and Il21r, which were colocalized with Cxcr4 in T cells . Notably, Gene Set Enrichment Analysis (GSEA) confirmed significant suppression of pathways related to cellular response to type II interferon and lymphocyte chemotaxis following antibody treatment . These findings establish a mechanistic foundation for CXCL12 antibody applications in inflammatory disease research.

What considerations are important when using CXCL12 antibodies as neutralizing agents in experimental models?

When employing CXCL12 antibodies as neutralizing agents in experimental models, researchers must address several critical considerations to ensure scientific rigor and reproducibility. First, antibody specificity must be thoroughly validated to confirm selective neutralization of CXCL12 without cross-reactivity to related chemokines. Second, appropriate dosing regimens must be established through dose-response studies, as therapeutic efficacy often follows a bell-shaped curve. The potency of neutralization depends on antibody affinity, which can vary between clones and lots; therefore, functional validation (e.g., migration assays) is recommended before initiating in vivo studies.

For experimental design, researchers should consider:

  • Route of administration (systemic vs. local) based on the target tissue and research question

  • Timing of intervention (preventive vs. therapeutic protocols)

  • Controls including isotype-matched non-specific antibodies

  • Potential immunogenicity of the antibody, particularly in long-term studies

  • Half-life and tissue distribution of the selected antibody

Recent studies have demonstrated that humanized CXCL12 antibodies can effectively modulate immune responses in disease models with minimal off-target effects . When analyzing CXCL12 antibody-specific DEGs, researchers found relatively few significant changes in biological processes unrelated to disease treatment, suggesting high specificity of therapeutic action . This favorable safety profile makes CXCL12 antibodies particularly valuable for in vivo experimental applications.

How can researchers distinguish between effects mediated through different CXCL12 receptors (CXCR4 vs. ACKR3) when using neutralizing antibodies?

Distinguishing between CXCR4-mediated and ACKR3-mediated effects following CXCL12 neutralization presents a significant challenge in experimental design. CXCL12 binds to both receptors, which can trigger distinct signaling pathways and cellular responses. To differentiate these effects, researchers should implement a multi-faceted approach combining complementary techniques.

First, researchers can employ receptor-specific antagonists alongside CXCL12 antibodies in parallel experimental groups. For example, AMD3100 (plerixafor) specifically blocks CXCR4 without affecting ACKR3, while CCX771 selectively inhibits ACKR3. By comparing phenotypes between CXCL12 antibody treatment alone versus combined with receptor-specific antagonists, researchers can parse receptor-specific contributions.

Second, genetic approaches using receptor-specific knockdown/knockout models provide definitive evidence for receptor involvement. Conditional or inducible systems are particularly valuable for avoiding developmental confounders. Researchers can neutralize CXCL12 in wild-type, CXCR4-deficient, and ACKR3-deficient backgrounds to determine receptor requirements.

Third, downstream signaling analysis helps differentiate receptor contributions. CXCR4 primarily signals through Gαi proteins activating MAPK and PI3K/Akt pathways, while ACKR3 predominantly functions as a scavenger receptor but can also signal through β-arrestin. Phospho-specific antibodies against ERK1/2, Akt, and other pathway components can identify active signaling cascades following treatment.

The CXCL12/CXCR4/ACKR3 axis represents a potential therapeutic target for inflammatory diseases through multiple mechanisms beyond simple cell migration inhibition . Understanding receptor-specific effects is crucial for interpreting experimental outcomes and developing targeted therapeutic strategies.

How should researchers interpret unexpected CXCL12 staining patterns in tissues previously unreported to express this chemokine?

When encountering unexpected CXCL12 staining in tissues not previously reported to express this chemokine, researchers should follow a systematic approach to validate findings rather than immediately dismissing the result as non-specific binding. First, researchers should consult comprehensive expression databases and recent literature to determine if the expression pattern has simply been underreported. For example, when researchers questioned CXCL12 staining in liver, Boster Scientific Support confirmed that "based on literature liver does express CXCL12" and cited Uniprot.org data showing CXCL12 expression across multiple tissues including liver .

To validate unexpected staining patterns, researchers should:

  • Perform technical validation using multiple antibody clones targeting different epitopes

  • Correlate protein detection with mRNA expression using RT-qPCR or in situ hybridization

  • Include appropriate positive and negative controls, including tissues known to express or lack CXCL12

  • Test antibody specificity through pre-absorption with recombinant CXCL12 protein

  • Consider cell type-specific expression patterns within heterogeneous tissues

Unexpected expression may indicate novel biological roles in specific tissues, developmental stages, or disease states. For example, fibroblast secretion of CXCL12 was confirmed as expected based on literature , highlighting the importance of cell type-specific analysis. Researchers should also consider post-translational modifications or splice variants that might affect antibody binding. Reporting novel expression patterns with thorough validation contributes valuable data to the scientific community.

What factors might contribute to variability in CXCL12 antibody performance between experimental replicates?

Variability in CXCL12 antibody performance between experimental replicates can stem from multiple factors that researchers should systematically address. First, antibody formulation variations between lots can significantly impact consistency. Some lots of CXCL12 antibodies may contain different stabilizers (e.g., BSA vs. BSA-free formulations), which can affect performance in specific applications . Researchers should record lot numbers and request consistency in formulation when possible.

Sample preparation techniques also critically influence reproducibility. CXCL12 can be sensitive to degradation during tissue processing, and its detection may be affected by fixation methods for immunohistochemistry or protein extraction protocols for Western blotting. Standardizing sample collection, storage, and processing helps minimize this variability.

Technical factors contributing to inconsistent results include:

  • Antibody concentration variations (accurate pipetting is essential)

  • Incubation time and temperature fluctuations

  • Washing stringency differences between experiments

  • Detection reagent variability or degradation

  • Different blocking reagents affecting background levels

Biological factors also contribute to variability. CXCL12 expression can be dynamically regulated by inflammatory stimuli, hypoxia, and circadian rhythms. Researchers should control for these variables by standardizing experimental timing and conditions. Additionally, tissue heterogeneity can lead to sampling variability, particularly in disease models where CXCL12 expression may be focal rather than uniform. Employing tissue microdissection or single-cell approaches can help address this issue.

How can researchers integrate and interpret CXCL12 antibody data with other -omics datasets for comprehensive pathway analysis?

Integrating CXCL12 antibody data with other -omics datasets enables comprehensive pathway analysis that extends beyond simple protein detection. This integration approach has proven valuable in recent studies examining CXCL12 antibody treatment effects in disease models. Researchers should first establish clear analytical frameworks that facilitate meaningful comparisons across datasets while accounting for different data types and scales.

For effective integration strategies, researchers can follow the approach demonstrated in recent CXCL12 antibody studies that combined single-cell RNA sequencing with antibody treatment data. This workflow involved:

  • Identifying cell populations affected by CXCL12 antibody treatment using scRNA-seq clustering

  • Performing differential expression analysis to identify genes modulated by antibody treatment

  • Conducting pseudobulk RNA-seq analysis from transcript counts

  • Applying network analysis (e.g., STRING) to identify protein-protein interaction networks

  • Performing community detection to identify major functional clusters

  • Conducting Gene Ontology enrichment and Gene Set Enrichment Analysis to identify affected pathways

This approach successfully identified 153 differentially expressed genes that increased in a disease model and decreased following antibody treatment, representing key mediators of both pathogenesis and therapeutic response . The methodology revealed that CXCL12 antibody treatment modulated genes involved in immune cell chemotaxis, chemokine signaling, and interferon responses, providing mechanistic insights into therapeutic effects .

When integrating antibody data with transcriptomics, researchers should be aware that protein and mRNA levels may not perfectly correlate due to post-transcriptional regulation. Including proteomics data provides additional validation. For comprehensive insights, researchers should consider temporal dynamics by collecting samples at multiple timepoints to capture the full trajectory of CXCL12-mediated responses following antibody administration.

How do humanized CXCL12 antibodies compare to small molecule CXCR4 antagonists in research and potential therapeutic applications?

Humanized CXCL12 antibodies and small molecule CXCR4 antagonists represent distinct approaches to targeting the CXCL12/CXCR4 axis with important differences in their research applications and therapeutic potential. Humanized CXCL12 antibodies directly neutralize the ligand, preventing its interaction with both CXCR4 and ACKR3 receptors. This approach offers high specificity for the target chemokine and potentially fewer off-target effects. Recent transcriptomic analysis of CXCL12 antibody-treated disease models revealed minimal changes in biological processes unrelated to the disease pathways, suggesting high target specificity .

Key comparative factors include:

  • Specificity: Antibodies typically offer higher specificity compared to small molecules

  • Half-life: Antibodies generally have longer half-lives, allowing less frequent dosing

  • Tissue penetration: Small molecules may penetrate tissues more effectively than antibodies

  • Administration route: Small molecules often allow oral administration, while antibodies typically require injection

  • Production complexity: Antibodies involve more complex manufacturing processes

Both approaches have demonstrated efficacy in delaying disease onset or preventing progression in models of cancer, viral infections, inflammatory bowel diseases, rheumatoid arthritis, and osteoarthritis . The choice between these therapeutic strategies should be guided by the specific research question, disease context, and practical considerations such as administration route and pharmacokinetic requirements.

What insights from single-cell RNA sequencing studies have expanded our understanding of CXCL12 antibody mechanisms in immune modulation?

Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of CXCL12 antibody mechanisms in immune modulation by providing unprecedented resolution of cellular responses. Recent scRNA-seq studies examining CXCL12 antibody treatment in inflammatory disease models have revealed complex cellular and molecular dynamics that extend far beyond simple chemotaxis inhibition.

In an alopecia areata (AA) mouse model, scRNA-seq analysis demonstrated that CXCL12 antibody treatment significantly altered immune cell proportions, particularly reducing the expanded CD8+ T cell and dendritic cell/macrophage populations characteristic of the disease state . Differential expression analysis identified 153 genes that were upregulated in the disease model and subsequently downregulated following antibody treatment, representing key mediators of both pathogenesis and therapeutic response .

Network analysis of these differentially expressed genes revealed three major functional clusters:

  • Cluster A: Immune cell chemotaxis (lymphocytes and monocytes), chemokine-mediated signaling, cellular response to type II interferon, and regulation of leukocyte differentiation

  • Cluster B: Complement system related to functions of dendritic cells and macrophages

  • Cluster C: Cytokine response pathways, including responses to type I and II interferons

Gene Set Enrichment Analysis further confirmed that pathways related to cellular response to type II interferon and lymphocyte chemotaxis were significantly elevated in the disease model and suppressed following antibody treatment . The study specifically identified key immune-related genes including Ifng, Cd8a, Ccr5, Ccl4, Ccl5, and Il21r, which were colocalized with Cxcr4 in T cells and regulated by CXCL12 antibody treatment .

These insights demonstrate that CXCL12 antibodies exert multifaceted effects on immune signaling networks rather than simply blocking chemotaxis, providing a more nuanced foundation for therapeutic applications.

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.