Recombinant Human Stromal cell-derived factor 1 protein (CXCL12) (Active)

Shipped with Ice Packs
In Stock

Description

Production and Quality Control

Recombinant CXCL12 is synthesized in E. coli with stringent purification protocols:

Functional Mechanisms

CXCL12 signals through G protein-coupled receptors CXCR4 and CXCR7, triggering diverse pathways:

Key Activities

  • Chemotaxis: Mobilizes T-cells, monocytes, and CD34+ hematopoietic stem cells

  • Cell Survival: Synergizes with IL-7, GM-CSF, and thrombopoietin to inhibit apoptosis

  • Receptor Internalization: Induces β-arrestin recruitment and CXCR4 downregulation

  • Heparin Binding: Stabilizes chemokine gradients and protects against proteolysis

Signaling Pathways

  1. Calcium Mobilization: Rapid intracellular Ca²⁺ flux via Gαi proteins

  2. ERK1/2 Phosphorylation: Activates MAPK pathways for proliferation/migration

  3. Integrin Activation: Enhances adhesion via CXCR4-independent binding to ITGAV:ITGB3

Experimental Findings

  • Hematopoiesis: Transgenic CXCL12 mice show elevated myeloid progenitor survival and splenic myelopoiesis .

  • HIV Inhibition: Competes with HIV-1 for CXCR4 binding, reducing viral entry .

  • Cancer Metastasis: Promotes tumor cell migration via CXCR4/ACKR3 interactions .

  • Tissue Repair: COAM (chlorite-oxidized oxyamylose) protects CXCL12 from MMP-9 cleavage, preserving activity in myocardial infarction models .

Functional Assay Data

AssayResultSource
Calcium MobilizationEC50 = 160 nM (THP-1 cells)
Chemotaxis (THP-1)100% migration at 10 ng/mL; abolished by MMP-9 cleavage
ERK1/2 ActivationPhosphorylation restored by COAM (10x–100x excess) post-MMP-9 treatment

Clinical and Therapeutic Relevance

  • Cardioprotection: Reduces infarct size post-myocardial infarction .

  • Neuroregeneration: Enhances neural progenitor survival in Alzheimer’s/Parkinson’s models .

  • Limitations: Susceptibility to proteolysis and species-specific activity (e.g., no effect on neutrophils) .

Challenges and Future Directions

  • Stability Optimization: Engineering protease-resistant mutants .

  • Delivery Systems: Nanoparticle encapsulation for sustained release .

  • Dual Targeting: Combining CXCL12 with checkpoint inhibitors in cancer immunotherapy .

Product Specs

Buffer
20 mM Phosphate Buffered Saline, pH 7.0, 130 mM Sodium Chloride
Form
Available as liquid solution or lyophilized powder.
Lead Time
5-10 business days
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations are stable for 6 months at -20°C/-80°C, while lyophilized preparations remain stable for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple uses, aliquot to avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
12-O-tetradecanoylphorbol 13-acetate repressed protein 1; AI174028; C-X-C motif chemokine 12; Chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); Chemokine (C-X-C motif) ligand 12; Chemokine CXC motif ligand 12; cxcl12; hIRH; hSDF-1; Intercrine reduced in hepatomas; IRH; OTTHUMP00000019491 ; PBSF; Pre-B cell growth-stimulating factor; SCYB12; SDF 1; SDF-1; SDF-1-alpha(3-67); SDF-1a; SDF-1b; SDF1_HUMAN; SDF1A; SDF1B; Stromal cell-derived factor 1; Stromal cell-derived factor 1 delta ; Stromal cell-derived factor 1 gamma ; Stromal cell-derived factor 1a ; Stromal cell-derived factor-1 alpha ; Thymic lymphoma cell-stimulating factor; Tlsf; TLSF-a; TLSF-b; Tlsfa; Tlsfb; TPAR1
Datasheet & Coa
Please contact us to get it.
Expression Region
22-89aa
Mol. Weight
8.0 kDa
Protein Length
Full Length of Mature Protein of Isoform Alpha
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Recombinant Human Stromal cell-derived factor 1 protein (CXCL12) is a chemoattractant primarily active on T-lymphocytes and monocytes, but not neutrophils. It activates the C-X-C chemokine receptor CXCR4, inducing a rapid and transient increase in intracellular calcium ion levels and subsequent chemotaxis. Truncated isoforms, such as SDF-1-beta(3-72) and SDF-1-alpha(3-67), exhibit reduced chemotactic activity. Binding to cell surface proteoglycans appears to inhibit the formation of inactive isoforms, thereby preserving activity at localized sites. CXCL12 also binds to the atypical chemokine receptor ACKR3, initiating beta-arrestin signaling and functioning as a scavenger receptor for SDF-1. Furthermore, CXCL12 interacts with 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. CXCL12 stimulates the migration of monocytes and T-lymphocytes through CXCR4 and ACKR3, while concurrently reducing monocyte adhesion to ICAM-1-coated surfaces. This anti-adhesive effect is mediated by the inhibition of CXCR4-mediated LFA-1 (beta-2 integrin) adhesion to ICAM-1 via LYN kinase. CXCL12 also inhibits CXCR4-mediated infection by T-cell line-adapted HIV-1. It plays a crucial protective role following myocardial infarction, and induces the downregulation and internalization of ACKR3 in various cells. CXCL12 is essential for several developmental processes, including B-cell lymphopoiesis, myelopoiesis in the bone marrow, and heart ventricular septum formation. It also stimulates the proliferation of bone marrow-derived B-cell progenitors in the presence of IL-7 and supports the growth of stromal cell-dependent pre-B-cells.
Gene References Into Functions
  • CXCL12-positive cases showed shorter disease-free survival compared to CXCL12-negative cases. PMID: 30182340
  • This study highlights the crucial role of CXCR7, along with CXCR4, in regulating the migration and homing of both normal and malignant hematopoietic cells in response to CXCL12. PMID: 29433559
  • CXCL12 rs1801157 is independently associated with Human papillomavirus infection and influences the development of high-grade intraepithelial lesions. PMID: 30227860
  • CXCL12 silencing showed a protective effect against podocyte injury, potentially through the inhibition of the CXCL12/STAT3 signaling pathway. PMID: 29508174
  • CXCL12/SDF1 protein expression is a favorable prognostic biomarker in breast cancer. PMID: 29800557
  • The CXCL12-CXCR4 axis promotes migration, invasion, and epithelial-mesenchymal transition (EMT) in B-CPAP cells, partly through activation of the NF-κB signaling pathway. PMID: 29316404
  • This study demonstrates methylation-mediated epigenetic regulation of CXCL12 gene expression in papillary thyroid carcinoma (PTC), suggesting a role for CXCL12 in PTC development. PMID: 28272462
  • Non-oxidizable HMGB1 induces sustained cardiac fibroblast migration by altering the CXCL12/CXCR4 axis, impacting cardiac remodeling post-infarction. PMID: 28716707
  • This research provides insights into the regulation of CXCL12 gene transcription via its 3'UTR and its implications in diseases associated with abnormal CXCL12α expression. PMID: 30266500
  • High SDF-1 expression correlates with bladder cancer progression. PMID: 30015971
  • High CXCL12 expression is associated with metastasis in colon cancer. PMID: 29305742
  • MiR-125b acts as a downstream mediator upon activation of the CXCL12/CXCR4 axis. PMID: 28176874
  • The CXCL12-related rs18011517 polymorphism is more prevalent in non-Hodgkin lymphoma patients and may be associated with disease pathogenesis and outcome. PMID: 30197351
  • CXCL12 and CXCR4 are crucial for maintaining homeostasis, particularly during hematopoiesis. Clinical trials are investigating the use of CXCR4 inhibitors to improve cancer treatment efficacy. PMID: 29288743
  • B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cells migrate towards mesenchymal stromal cells (MSCs) in a CXCL12-dependent manner. PMID: 28619846
  • Elevated serum CXCR4 and CXCL12 levels in septic neonates suggest their potential as diagnostic biomarkers for neonatal sepsis. PMID: 28562124
  • CXCL12 and CXCR4 polymorphisms may be risk factors and potential markers for hepatocellular carcinoma (HCC). PMID: 29741398
  • The SDF-1/CXCR4 axis induces human dental pulp stem cell migration via FAK/PI3K/Akt and GSK3β/β-catenin pathways. PMID: 28067275
  • EGFR overexpression and mutations alter the biological characteristics of human lung adenocarcinoma cells through the CXCR4/CXCL12 signaling pathway. PMID: 30037369
  • Serum CXCL12, but not CXCR4, is associated with head and neck squamous cell carcinomas. PMID: 29693336
  • This study investigated the expression of fibrosis markers, ERα, and the SDF-1/CXCR4 axis in intrauterine adhesions endometrium. PMID: 29568895
  • HIV-1 infected individuals with the SDF-1 3'A polymorphism have a higher risk of developing late-stage AIDS. PMID: 30053458
  • The SDF1/CXCR4 signaling pathway is involved in low-intensity pulsed ultrasound-promoted periodontal ligament stem cell migration. PMID: 29620151
  • SDF-1 can rapidly activate integrins allosterically by binding to site 2, independent of CXCR4, representing a novel drug target. PMID: 29301984
  • Association between CXCL12 single nucleotide polymorphisms and hypertension risk in the Chinese Han population. PMID: 30180964
  • The CXCR4-CXCL12 chemokine axis plays a critical role in breast cancer progression and prognosis. PMID: 29516917
  • Elevated serum SDF-1 levels are associated with primary biliary cholangitis and may serve as a potential marker. PMID: 29414663
  • The CXCR4/CXCL12 axis disruption by the CXCR4 antagonist AMD3100 inhibited metastasis to the liver. PMID: 29436696
  • SDF-1α overexpression in bone marrow-derived stromal stem cells promotes bone formation via osteogenesis and angiogenesis. PMID: 29758548
  • SDF-1, as an inflammatory cytokine, induces MMP expression in human endplate chondrocytes, potentially influencing stem cell homing for nucleus pulposus regeneration. PMID: 29207021
  • Hypoxia does not affect the angiogenic capacity of endothelial progenitor cells (EPCs), and VEGF-A and SDF-1 exhibit compensatory angiogenic effects. PMID: 27943613
  • Estrogen may promote ER-negative breast cancer progression by stimulating SDF-1α secretion from cancer-associated fibroblasts, recruiting myeloid-derived suppressor cells (MDSCs) to the tumor microenvironment. PMID: 27996037
  • SDF-1 and CXCR4 expression are important for loco-regional control and overall survival in head and neck squamous cell carcinoma (HNSCC) after radiochemotherapy. PMID: 29061496
  • Adipocyte autocrine SDF-1 function regulates insulin resistance; SDF-1 gene expression correlates with insulin resistance in adipocytes. PMID: 29581126
  • SDF-1α stimulates E-selectin/ligand expression in endothelial cells and bone marrow-derived endothelial progenitor cells (EPCs), enhancing EPC-EC interactions in ischemic wounds. PMID: 27713493
  • CXCL12(5-67) does not promote neural stem cell (NSC) migration but induces cell death. PMID: 28623786
  • A SDF-1/CXCR4-RhoA and RhoC-ROS-cytoskeleton pathway regulates Jurkat cell migration in response to SDF-1. PMID: 28536953
  • Upregulation of miR31 expression was validated using GEO datasets. PMID: 27597234
  • Differential expression of SDF-1 receptor CXCR4 in inherited thrombocytopenias. PMID: 28032520
  • Review article on the role of CXCL12 in bladder cancer. PMID: 29022185
  • Intravenous rhSDF-1α accelerates re-endothelialization in aneurysm necks after flow diverter implantation. PMID: 28159982
  • Increased invasiveness at MELF pattern sites potentially through CXCL14-CXCR4 and CXCL12-CXCR4 axes. PMID: 28277316
  • SDF-1 rs1801157 polymorphism may be a risk factor for cancer development, particularly urologic and lung cancers, in Asian populations. PMID: 27265091
  • SDF1 polymorphism shows a moderate protective effect against AIDS progression in specific populations. PMID: 29420545
  • EMT, migration, and survival in MCF-7 and H460 cells depend on the release of FHC control on iron/ROS metabolism and the CXCR4/CXCL12 axis. PMID: 28774348
  • Higher serum levels in preeclamptic women. PMID: 28001450
  • A defect in CXCL12 promoter histone acetylation may contribute to CXCL12 expression extinction in colon cancer. PMID: 28418886
  • The SDF-1α/CXCR4 signaling pathway is associated with clinicopathological features and prognosis in nasopharyngeal carcinoma. PMID: 28559386
  • The CXCL12-CXCR7 axis accelerates pancreatic cancer cell migration and invasion via mTOR and Rho/ROCK pathways and predicts poor prognosis. PMID: 27542220
  • Review article on 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 are the structural characteristics of human CXCL12/SDF-1?

CXCL12/SDF-1 is a member of the CXC (or alpha) family of chemokines. The protein exhibits a typical three antiparallel beta-strand chemokine-like fold. Human CXCL12 is expressed as five different isoforms (α, β, γ, δ, ε) that differ only in their C-terminal tails but share the same core structure . Specifically, the gamma isoform is a 12 kDa, heparin-binding protein. Human SDF-1 gamma is synthesized as a 119 amino acid precursor containing a 21 amino acid signal sequence followed by a 98 amino acid mature region .

Notably, mature SDF-1 molecules are not glycosylated. The N-terminal amino acids 1-8 form a receptor binding site, while amino acids 1 and 2 (Lys-Pro) are specifically involved in receptor activation . This structural configuration is critical for the protein's biological activity.

What is the biochemical equilibrium state of recombinant CXCL12/SDF-1 in solution?

Biophysical characterization using multiple techniques has demonstrated that SDF-1α exists in a monomer-dimer equilibrium in solution. Initial indications of self-association were observed through static light scattering measurements of the average mass. This was confirmed through sedimentation velocity ultracentrifugation, which identified two distinct species corresponding to monomer and dimer forms .

Further analysis using sedimentation equilibrium ultracentrifugation and dynamic light scattering yielded a composite dimerization constant (Kd) of 150 ± 30 μM . This equilibrium is an important consideration for researchers designing experiments, as the monomer-dimer ratio may influence biological activity and interaction with receptors.

How does CXCL12/SDF-1 signal through its receptor and what are the immediate downstream effects?

CXCL12/SDF-1 primarily signals through the G protein-coupled receptor CXCR4. Upon binding, the N-terminal region of CXCL12 activates CXCR4, which is expressed on multiple cell types including leukocytes, hematopoietic stem cells, and other progenitor cells . This interaction triggers several downstream signaling cascades.

The signaling process can be monitored through several methodological approaches:

  • Intracellular calcium mobilization: CXCL12 binding to CXCR4 induces a rapid and measurable increase in intracellular calcium levels .

  • MAP kinase activation: The interaction leads to phosphorylation of ERK1/2, which can be detected by Western blotting .

  • Receptor internalization: Upon activation, CXCR4 undergoes internalization, which can be quantified using flow cytometry to measure cell surface CXCR4 levels before and after CXCL12 stimulation .

  • Chemotaxis: Functional CXCL12-CXCR4 signaling results in directional cell migration that can be measured in transwell migration assays .

These methodologies provide complementary approaches to assess CXCL12 activity in experimental settings.

What are the primary biological functions of CXCL12/SDF-1 in hematopoiesis?

CXCL12/SDF-1 plays crucial roles in hematopoiesis, particularly in the survival and function of hematopoietic stem and progenitor cells. The protein directly enhances survival and prevents apoptosis of myeloid progenitor cells through CXCR4 and G(alpha)i protein-mediated signaling .

This effect has been demonstrated on multiple types of progenitor cells, including human bone marrow (BM) and cord blood (CB) colony-forming units-granulocyte macrophage (CFU-GM), burst-forming units-erythroid (BFU-E), and CFU-granulocyte-erythroid-macrophage-megakaryocyte (CFU-GEMM) . The direct-acting nature of this effect has been confirmed through single-cell assays with isolated CD34+++ cells.

Additionally, CXCL12 significantly enhances the engrafting capability of mouse long-term, marrow-competitive, repopulating stem cells when cultured ex vivo with other growth factors like interleukin-6 and steel factor . This function has important implications for ex vivo expansion protocols and gene-transduction procedures in research and clinical applications.

How can researchers effectively produce and purify recombinant CXCL12/SDF-1 for functional studies?

Recombinant production of CXCL12/SDF-1 can be accomplished using Escherichia coli expression systems. A documented protocol involves producing the chemokine as methionine-SDF-1α in E. coli without the leader peptide sequence . The protein undergoes a denaturation and refolding process, followed by purification via reversed-phase HPLC.

The critical steps in this methodology include:

  • Expression optimization: Yields of 1-2 mg of purified protein per gram of wet cell paste have been reported .

  • Purity assessment: SDS-PAGE analysis should confirm >95% purity of the final product .

  • Functional validation: The refolded protein should be tested for CXCR4 binding capacity, as properly refolded CXCL12 functions as a ligand for CXCR4 receptor and demonstrates the ability to block HIV-mediated cell fusion and downmodulate CXCR4 receptors .

This methodology enables production of sufficient quantities (hundreds of milligrams) of biologically active protein for detailed structural and functional studies.

What experimental methods can be used to assess CXCL12/SDF-1 proteolytic processing and its impact on function?

CXCL12/SDF-1 is subject to proteolytic processing that significantly affects its biological activity. Two key proteases known to cleave CXCL12 are matrix metalloproteinase-9 (MMP-9) and dipeptidyl peptidase-4 (CD26) . Researchers can employ several methodologies to assess this processing:

  • Western blot analysis: Using antibodies that specifically recognize either intact or all forms of CXCL12. A monoclonal antibody detecting only the intact form (visualized with green fluorescence) can be used alongside a polyclonal antibody recognizing both cleaved and intact forms (visualized with red fluorescence) .

  • Functional assays to compare intact versus proteolytically processed CXCL12:

    • Intracellular calcium mobilization assays

    • ERK1/2 phosphorylation by Western blotting

    • CXCR4 receptor internalization measured by flow cytometry

    • Chemotaxis assays using Transwell systems

The data below illustrates how proteolytic processing affects CXCL12 activity:

Functional AssayIntact CXCL12MMP-9 Cleaved CXCL12CD26 Cleaved CXCL12
Calcium signalingActive (100%)Inactive (0%)Inactive (0%)
ERK1/2 phosphorylationActive (100%)Inactive (0%)Reduced
CXCR4 internalization~25% reductionNo effect (0%)Reduced
ChemotaxisActive (100%)Inactive (0%)Reduced

These methodologies provide comprehensive assessment of proteolytic processing effects on CXCL12 function.

How can researchers protect CXCL12/SDF-1 from proteolytic inactivation during experiments?

Protecting CXCL12/SDF-1 from proteolytic degradation is crucial for maintaining its biological activity in experimental settings. Research has identified chlorite-oxidized oxyamylose (COAM) as an effective protectant against proteolytic inactivation by both MMP-9 and CD26 .

The methodology for implementing this protection involves:

  • Pre-incubation: CXCL12 should be pre-incubated with COAM before exposure to proteases. A mass excess ratio of 100× COAM to CXCL12 provides optimal protection (approximately 80% of intact form remaining after protease exposure) .

  • Validation of protection: Western blot analysis using antibodies specific for the intact form can confirm the protective effect .

  • Functional verification: Protected CXCL12 should be tested in functional assays (calcium signaling, ERK1/2 phosphorylation, receptor internalization, or chemotaxis) to confirm that biological activity is maintained .

Even intermediate concentrations (10× mass excess) of COAM provide significant, though partial, protection of CXCL12 activity, making this approach adaptable to various experimental constraints .

What are the appropriate protocols for testing CXCL12/SDF-1 effects on myeloid progenitor cell survival?

To assess CXCL12/SDF-1 effects on myeloid progenitor cell survival, researchers can employ colony formation assays with appropriate dose-response curves. Studies have demonstrated that CXCL12 has survival-enhancing and antiapoptotic effects on various progenitor populations .

The methodology involves:

  • Cell isolation: Obtain human bone marrow, cord blood, or mouse bone marrow cells, with specific focus on colony-forming units (CFU)-granulocyte macrophage, burst-forming units-erythroid, and CFU-granulocyte-erythroid-macrophage-megakaryocyte .

  • Single cell assays: For direct-acting effects, isolate CD34+++ cells and plate them individually with appropriate growth factors with and without CXCL12 .

  • Dose determination: The effective dose (ED50) for CXCL12 effects ranges from 4-24 ng/mL, providing a starting point for dose-response experiments .

  • Receptor involvement: Include experimental conditions with CXCR4 antagonists and G(alpha)i protein inhibitors to confirm the mechanism of action .

  • Ex vivo culture assessment: For stem cell engraftment studies, culture cells with CXCL12 along with other growth factors (e.g., interleukin-6 and steel factor) for 48 hours before competitive repopulation assays .

This comprehensive approach allows for detailed characterization of CXCL12's effects on hematopoietic cell survival and function.

How can researchers distinguish between different CXCL12/SDF-1 isoforms in experimental systems?

CXCL12/SDF-1 exists in five isoforms (α, β, γ, δ, ε) that differ in their C-terminal tails . Distinguishing between these isoforms requires specific methodological approaches:

  • Molecular identification: PCR-based methods using isoform-specific primers targeting the unique C-terminal sequences can differentiate between isoforms at the mRNA level.

  • Protein detection: Western blotting with antibodies recognizing common regions will detect all isoforms, while antibodies targeting the unique C-terminal regions can be used for isoform-specific detection. For the γ isoform, antibodies recognizing the unique region of human CXCL12 (Lys22-Asn119, Accession # NP_001029058) would be suitable .

  • Functional characterization: The isoforms may exhibit different biological activities due to their distinct C-terminal regions. Systematic comparative analysis of their effects on calcium signaling, ERK phosphorylation, and chemotaxis can reveal functional differences.

  • Interaction studies: The isoforms may differ in their ability to bind heparin and other glycosaminoglycans due to variations in their C-terminal regions. Heparin-binding assays can help distinguish between isoforms with different affinities for glycosaminoglycans.

These approaches enable researchers to identify and characterize specific CXCL12 isoforms in their experimental systems.

What controls should be included when assessing CXCL12/SDF-1 proteolytic processing in experimental systems?

When studying CXCL12/SDF-1 proteolytic processing, researchers should include the following controls to ensure reliable and interpretable results:

  • Intact protein control: Unprocessed CXCL12 should be included as a positive control for maximum activity in all functional assays .

  • Protease-only control: Samples containing only the protease (MMP-9 or CD26) should be included to confirm protease activity and specificity .

  • Protease inhibitor controls: Specific inhibitors of MMP-9 or CD26 should be included to confirm that the observed effects are due to the specific protease activity rather than contaminants .

  • Antibody validation: When using antibodies to detect intact versus cleaved forms, controls should include samples with known cleavage status to confirm antibody specificity .

  • Functional readout controls: For calcium signaling, ERK phosphorylation, receptor internalization, or chemotaxis assays, include positive controls (intact CXCL12) and negative controls (buffer only) to establish the dynamic range of the assay .

  • Concentration-matched controls: When comparing different forms or treatments of CXCL12, ensure that equimolar concentrations are used to allow direct comparison of activity levels .

Including these controls ensures that the experimental system is functioning properly and that the observed effects are specifically related to CXCL12 proteolytic processing.

How should researchers interpret contradictory results between different functional assays of CXCL12/SDF-1 activity?

When encountering contradictory results between different functional assays of CXCL12/SDF-1 activity, researchers should consider several factors in their interpretation:

  • Assay sensitivity differences: The various assays (calcium signaling, ERK phosphorylation, receptor internalization, chemotaxis) have different sensitivity thresholds. For instance, partially processed CXCL12 may retain sufficient activity to trigger ERK phosphorylation but not chemotaxis .

  • Temporal dynamics: The assays measure events occurring at different time points in the signaling cascade. Calcium signaling occurs within seconds to minutes, ERK phosphorylation within minutes, receptor internalization within 15-30 minutes, and chemotaxis over hours .

  • Receptor density effects: Cell lines or primary cells used in different assays may express varying levels of CXCR4, affecting response magnitude. Flow cytometry should be used to quantify receptor expression across experimental systems .

  • Signaling pathway bifurcation: CXCR4 activation triggers multiple downstream pathways that may be differentially affected by partial CXCL12 processing or experimental conditions.

  • Protein aggregation state: As CXCL12 exists in a monomer-dimer equilibrium (Kd = 150 ± 30 μM) , concentration differences between assays may alter this equilibrium and affect activity.

When contradictions arise, researchers should systematically investigate these factors, potentially using dose-response curves across all assays to identify threshold effects or pathway-specific sensitivities.

What are the critical considerations when translating in vitro findings about CXCL12/SDF-1 to in vivo applications?

Translating in vitro findings about CXCL12/SDF-1 to in vivo applications requires careful consideration of several factors:

  • Proteolytic environment: In vivo, CXCL12 encounters various proteases including CD26 and MMP-9 that can cleave and inactivate it . Protective strategies such as COAM pre-treatment or protease-resistant CXCL12 variants should be considered for in vivo applications .

  • Context-dependent effects: CXCL12 can have beneficial or detrimental effects depending on the organ system or disease context . The same signaling pathway may promote tissue repair in one context but exacerbate pathology in another.

  • Glycosaminoglycan interactions: In vivo, CXCL12 binds to glycosaminoglycans, which affects its localization, concentration gradient formation, and receptor activation . This interaction, absent in many in vitro systems, is critical for proper in vivo function.

  • Isoform-specific effects: The five isoforms of CXCL12 may have distinct in vivo functions related to their different C-terminal domains . The isoform used in vitro may not represent the predominant or most relevant isoform in the target tissue.

  • Pharmacokinetics: In vivo applications must account for distribution, metabolism, and clearance of administered CXCL12, which are not factors in most in vitro systems.

  • Cell-type specific responses: While in vitro studies often use homogeneous cell populations, in vivo applications involve diverse cell types with varying levels of CXCR4 expression and downstream signaling capabilities .

Researchers should design in vivo experiments with these considerations in mind, potentially incorporating proteolytic protection strategies and isoform-specific approaches based on the target tissue context.

How can CXCL12/SDF-1 be utilized in ex vivo expansion protocols for hematopoietic stem cells?

CXCL12/SDF-1 has significant potential for enhancing ex vivo expansion protocols for hematopoietic stem cells (HSCs), as it demonstrates direct survival-enhancing and antiapoptotic effects on these cells . Implementing CXCL12 in such protocols requires careful methodological considerations:

  • Combinatorial approach: CXCL12 should be used in combination with other growth factors. Research has demonstrated enhanced engraftment capability when mouse long-term, marrow-competitive, repopulating stem cells were cultured with CXCL12 alongside interleukin-6 and steel factor for 48 hours .

  • Dose optimization: The effective dose range for CXCL12 effects on myeloid progenitors is 4-24 ng/mL , providing a starting point for optimization in HSC expansion protocols.

  • Proteolytic protection: To maintain CXCL12 activity during the prolonged culture period required for expansion, researchers should consider protective strategies such as COAM pre-treatment or regular supplementation of fresh CXCL12.

  • Monitoring stem cell characteristics: Throughout the expansion protocol, researchers should monitor not only cell numbers but also the maintenance of stem cell phenotype (through flow cytometry for stem cell markers) and function (through colony formation and in vivo repopulation assays) .

  • Temporal considerations: The timing of CXCL12 addition may be critical, as its effects may differ depending on the stage of ex vivo culture and the activation state of the stem cells.

This approach leverages CXCL12's biological properties to enhance both the quantity and quality of HSCs in ex vivo expansion systems.

What methodological approaches can be used to study the interaction between CXCL12/SDF-1 and glycosaminoglycans?

The interaction between CXCL12/SDF-1 and glycosaminoglycans (GAGs) is critical for its in vivo function . Researchers can employ several methodological approaches to study this interaction:

  • Solid-phase binding assays: Immobilize different GAGs (heparin, heparan sulfate, chondroitin sulfate) on plates and measure binding of labeled CXCL12, with competition assays to determine binding specificity.

  • Surface plasmon resonance (SPR): Determine binding kinetics and affinity constants between CXCL12 and various GAGs in real-time using platforms like Biacore.

  • Isothermal titration calorimetry (ITC): Measure the thermodynamic parameters of CXCL12-GAG interactions, providing insights into the energetics of binding.

  • Nuclear magnetic resonance (NMR) spectroscopy: Identify specific CXCL12 residues involved in GAG binding through chemical shift perturbation experiments.

  • Functional assays with GAG competition: Assess how soluble GAGs affect CXCL12-induced calcium signaling, ERK phosphorylation, or chemotaxis to understand the functional consequences of GAG binding.

  • Mutagenesis studies: Create CXCL12 variants with mutations in putative GAG-binding regions to identify essential residues for this interaction.

  • In vivo localization studies: Compare the tissue distribution of wild-type CXCL12 versus GAG-binding deficient mutants to understand how this interaction affects in vivo localization.

These approaches provide complementary information about the structural and functional aspects of CXCL12-GAG interactions, essential for understanding gradient formation and tissue-specific activities.

How does the monomer-dimer equilibrium of CXCL12/SDF-1 affect its biological activity, and how can this be experimentally addressed?

The monomer-dimer equilibrium of CXCL12/SDF-1 (Kd = 150 ± 30 μM) potentially affects its biological activity through altered receptor binding kinetics, GAG interactions, and resistance to proteolytic processing. This phenomenon can be experimentally addressed through several approaches:

  • Concentration-dependent functional assays: Perform dose-response studies across a wide concentration range (spanning below and above the Kd) for calcium signaling, ERK phosphorylation, and chemotaxis to identify potential differences in activity between predominantly monomeric and dimeric states .

  • Engineered monomer/dimer variants: Create disulfide-locked dimers or monomeric variants (through mutations at the dimer interface) to directly compare the activities of these fixed states.

  • Real-time analysis of equilibrium: Use techniques like analytical ultracentrifugation or dynamic light scattering to monitor the monomer-dimer ratio under various experimental conditions that mimic physiological environments .

  • Differential susceptibility to proteases: Compare the sensitivity of monomeric versus dimeric CXCL12 to proteolytic processing by MMP-9 and CD26 to determine if dimerization affects proteolytic resistance .

  • Receptor binding studies: Use surface plasmon resonance or fluorescence-based binding assays to compare the receptor binding properties of monomeric versus dimeric CXCL12.

  • GAG interaction analysis: Determine whether monomeric and dimeric CXCL12 differ in their affinity for various GAGs, which could affect gradient formation and in vivo activity.

These experimental approaches would provide valuable insights into how the quaternary structure of CXCL12 influences its diverse biological functions and could guide the development of variants with enhanced stability or activity for research and therapeutic applications.

What are the current methodological challenges in studying CXCL12/SDF-1 protective effects in tissue repair models?

Studying CXCL12/SDF-1 protective effects in tissue repair models presents several methodological challenges that researchers must address:

  • Proteolytic inactivation: In injury environments, increased protease activity can rapidly degrade CXCL12, limiting its effectiveness. Researchers should consider using protease-resistant variants or protective strategies like COAM pre-treatment .

  • Delivery method optimization: Determining the optimal delivery method (direct injection, controlled release systems, gene therapy) and dosing regimen to achieve sustained, localized CXCL12 activity remains challenging.

  • Isoform selection: The five CXCL12 isoforms may have distinct activities in tissue repair. Systematic comparison of isoform-specific effects is necessary but methodologically complex, requiring isoform-specific reagents and detection methods .

  • Context-dependent effects: CXCL12 can have beneficial effects on tissue repair in some contexts but potentially harmful effects in others . Carefully designed experimental controls are needed to distinguish these outcomes.

  • Cell recruitment versus direct effects: Determining whether observed benefits result from cell recruitment to the injury site or from direct effects on resident cells requires sophisticated lineage tracing and conditional knockout approaches.

  • Temporal dynamics: The timing of CXCL12 administration relative to injury is likely critical, necessitating time-course studies with multiple experimental groups.

  • Long-term versus short-term outcomes: Short-term improvements in tissue parameters may not translate to long-term functional benefits, requiring extended follow-up periods in experimental models.

Addressing these challenges requires multidisciplinary approaches combining protein engineering, controlled delivery systems, and sophisticated in vivo imaging to fully characterize CXCL12's protective effects in tissue repair.

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.