SDF 1g Human

Stromal Cell-Derived Factor-1 gamma Human Recombinant (CXCL12)
Shipped with Ice Packs
In Stock

Description

Gene Structure and Expression Dynamics

The CXCL12 gene resides on human chromosome 10 and produces three primary isoforms via alternative splicing:

  • SDF-1α (CXCL12a)

  • SDF-1β (CXCL12b)

  • SDF-1γ (CXCL12γ)

In postnatal nervous system development, SDF-1γ mRNA expression inversely correlates with SDF-1β:

  • Embryonic stages: SDF-1β dominates in nerve tissues .

  • Adulthood: SDF-1γ becomes predominant, suggesting a role in mature neural maintenance .

  • Peripheral nerve injury: Transient upregulation of SDF-1β occurs, while SDF-1γ levels remain stable .

Functional Roles in the Nervous System

SDF-1γ exhibits specialized functions in neural environments:

  • Neuronal migration: Facilitates chemotaxis of neuronal precursors during development .

  • Schwann cell signaling: Collaborates with neurons to regulate myelination and repair processes .

  • Neuroprotection: Potential cleavage into neuropeptides with roles in inflammation and survival .

Expression and Localization

  • Cellular sources: Neurons and Schwann cells are primary producers in the nervous system .

  • mRNA dynamics: SDF-1γ expression increases postnatally, peaking in adulthood .

Mechanistic Insights

  • Proteolytic processing: Computational models predict cleavage sites in SDF-1γ that may generate bioactive peptides, unlike SDF-1α/β .

  • Receptor interactions: Binds CXCR4 and CXCR7, similar to other isoforms, but with distinct downstream effects due to structural variations .

Disease Implications

  • Chronic wounds: Dysregulated SDF-1/CXCR4/CXCR7 pathways impair stem cell homing, though γ-specific data remain limited .

  • Neurodegeneration: Hypothesized to modulate neuroinflammation, but human studies are lacking .

Comparative Analysis of SDF-1 Isoforms

FeatureSDF-1αSDF-1βSDF-1γ
Primary FunctionHematopoiesis, angiogenesis Immune cell trafficking Neural development
Expression PeakUbiquitousEmbryonic stagesAdulthood
Therapeutic TargetingHIV entry inhibition Cancer metastasis Neuroregeneration (proposed)

Limitations and Future Directions

  • Human data scarcity: Most findings derive from rodent models; human SDF-1γ remains poorly characterized .

  • Functional assays: No direct evidence of SDF-1γ’s chemotactic or signaling activity in human cells exists.

  • Therapeutic potential: Requires validation in models of neural injury or degenerative diseases.

Product Specs

Introduction
Stromal cell-derived factor-1 (SDF-1), also known as Chemokine (C-X-C motif) ligand 12 (CXCL12), is a chemokine that plays a crucial role in directing cell migration and coordinating cellular processes. Existing in two forms, SDF-1a/CXCL12a and SDF-1b/CXCL12b, this signaling molecule is highly chemotactic for lymphocytes, influencing their movement within the body. During development, SDF-1 is vital for guiding hematopoietic cells from the fetal liver to the bone marrow, highlighting its importance in immune system development. Mice lacking the CXCL12 gene exhibit lethality either before or shortly after birth, underscoring the critical role of this chemokine. Beyond its role in immune cell development, SDF-1 also impacts neuronal function by modulating the electrical activity of nerve cells. Widely expressed in various tissues such as the brain, thymus, heart, lungs, liver, kidneys, spleen, and bone marrow, SDF-1 exerts its effects by binding to its receptor, CXCR4, which was previously termed fusin. While this interaction was previously considered highly specific, recent findings suggest that SDF-1 may also engage with the CXCR7 receptor. The genes encoding both CXCL12 and CXCR4 exhibit a high degree of conservation between humans and mice, indicating their evolutionary significance. In humans, the CXCL12 gene resides on chromosome 10, while in both humans and mice, the sequences of CXCL12 and CXCR4 share remarkable similarity, with 99% and 90% identity, respectively.
Description
SDF-1 gamma Human Recombinant, a product of E.coli expression, is a single, non-glycosylated polypeptide chain composed of 99 amino acids. With a molecular weight of 11.6kDa, this purified protein is obtained through proprietary chromatographic techniques.
Physical Appearance
White, sterile-filtered powder obtained through lyophilization.
Formulation
SDF-1g protein undergoes lyophilization from a concentrated solution (0.2µm filtered) in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
To reconstitute lyophilized SDF-1g, it is recommended to dissolve it in sterile 18M-cm H₂O at a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized SDF-1g remains stable at room temperature for up to 3 weeks. However, for extended storage, it is recommended to store it desiccated at temperatures below -18°C. After reconstitution, SDF-1 gamma should be stored at 4°C for 2-7 days. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
Exceeds 96.0% purity as determined by Reverse Phase High Performance Liquid Chromatography (RP-HPLC) and Sodium Dodecyl Sulphate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analyses.
Biological Activity
The biological activity, as measured by a chemotaxis bioassay utilizing PHA and rHuIL-2 activated human peripheral blood T-lymphocytes, falls within the concentration range of 30-100 ng/ml.
Synonyms
Stromal cell-derived factor 1, SDF-1, hSDF-1, C-X-C motif chemokine 12, Intercrine reduced in hepatomas, IRH, hIRH, Pre-B cell growth-stimulating factor, PBSF, CXCL12, SDF1, hSDF-1gamma, SDF-1g, TLSF, TPAR1, SCYB12.
Source
Escherichia Coli.
Amino Acid Sequence
GKPVSLSYRC PCRFFESHVA RANVKHLKIL NTPNCALQIV ARLKNNNRQV CIDPKLKWIQ EYLEKALNKG RREEKVGKKE KIGKKKRQKK RKAAQKRKN.

Q&A

What is SDF-1g and how does it differ from other SDF-1 isoforms?

SDF-1g (also known as CXCL12 gamma) is a 12 kDa heparin-binding chemokine belonging to the CXC family. Human SDF-1g is synthesized as a 119 amino acid precursor containing a 21 amino acid signal sequence and a 98 amino acid mature region. The most distinctive feature of SDF-1g compared to other isoforms is its unique C-terminal tail, which consists of 26 amino acids that are highly charged and contain four BBXB motifs (where B = basic amino acid, X = any amino acid). In contrast, the more prevalent SDF-1 alpha has only 4 unique C-terminal amino acids and binds heparin via a shared BBXB site located more N-terminally. This structural difference significantly impacts the binding properties and functions of SDF-1g compared to other isoforms.

The N-terminal amino acids 1-8 of all SDF-1 isoforms form a receptor binding site, with amino acids 1 and 2 (Lys-Pro) specifically involved in receptor activation. All SDF-1 molecules can undergo proteolytic processing of these first two N-terminal amino acids by CD26, which is thought to generate a reduced-activity chemokine variant.

What receptors does SDF-1g interact with and what cellular responses does it mediate?

SDF-1g primarily interacts with two G protein-coupled receptors: CXCR4 and CXCR7. Additionally, it can bind to syndecan-4, a cell surface proteoglycan. The interaction with these receptors mediates various cellular responses including chemotaxis, cell survival, and proliferation.

SDF-1g, like other SDF-1 isoforms, influences lymphopoiesis, enhances the survival of myeloid progenitor cells, regulates the patterning and cell number of neural progenitors, and promotes angiogenesis. The specificity of SDF-1-mediated effects can be demonstrated through neutralization with anti-SDF-1 antibodies or by using CXCR4 antagonists such as AMD 3100, which dose-dependently inhibit SDF-1 signaling.

How can researchers detect SDF-1g activity in endothelial cell models?

Researchers can assess SDF-1g activity in endothelial cells through multiple complementary approaches:

  • Gene expression analysis: Northern blot analysis can be used to detect SDF-1-induced HO-1 mRNA expression. Typically, treatment with 100 ng/ml (14 nM) SDF-1 shows time-dependent induction of HO-1 mRNA, with maximal expression at 4 hours post-treatment. Western blotting can confirm the corresponding increase in HO-1 protein levels, which typically peaks at 16 hours after SDF-1 exposure.

  • Functional angiogenesis assays:

    • Tube formation assay: Endothelial cells seeded on Matrigel will form tube-like structures in response to SDF-1g, which can be quantified microscopically.

    • Migration assay: Transwell or wound healing assays can measure the migratory response of endothelial cells to SDF-1g gradients.

    • Aortic ring assay: This ex vivo model involves culturing aortic segments on Matrigel and treating with SDF-1g (typically 100 ng/ml). Capillary sprouting can be observed and quantified after 5 days. The endothelial nature of these sprouts can be confirmed by metabolic uptake of DiI-Ac-LDL.

  • Receptor expression analysis: Flow cytometry can be used to determine the percentage of cells expressing CXCR4, the primary receptor for SDF-1. Studies have shown that approximately 38.7% of human aortic endothelial cells (HAECs) express CXCR4.

What methods are available for detecting SDF-1 gene polymorphisms in clinical samples?

SDF-1 gene polymorphisms, such as SDF-1 G801A (rs1801157), can be detected using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. The protocol involves:

  • Sample collection: Collect 2 mL of blood in EDTA tubes for genomic DNA extraction.

  • DNA extraction: Use commercial kits such as Gene JET Whole Blood Genomic DNA Purification Mini Kit.

  • PCR amplification:

    • Use specific primers: Forward primer (5′ CAG TCA ACC TGG GCA AAG CC 3′) and Reverse primer (5′ AGC TTT GGT CCT GAG AGT CC 3′)

    • Program the thermal cycler with an initial activation step at 95°C for 10 minutes, followed by denaturation at 95°C for 1 minute, and a final extension at 72°C for 10 minutes.

  • Restriction enzyme digestion: Digest PCR products at 37°C with 10U MspI enzyme.

  • Genotype interpretation:

    • GG wild type (homozygous for restriction site): Two fragments of 202-bp and 100-bp

    • AA (homozygous for absence of restriction site): Single 302-bp band

    • AG (heterozygous): Three bands of 302-bp, 202-bp, and 100-bp

How does SDF-1g promote angiogenesis through HO-1 signaling?

SDF-1g promotes angiogenesis through a heme oxygenase-1 (HO-1) dependent mechanism. The signaling cascade involves:

  • HO-1 induction: SDF-1g binding to CXCR4 leads to dose-dependent induction of HO-1 mRNA and protein expression in endothelial cells. Treatment with 100 ng/ml SDF-1 results in approximately six-fold induction of HO-1 mRNA, while 200 ng/ml produces about eight-fold induction.

  • Protein kinase C (PKC) ζ activation: SDF-1g-mediated HO-1 induction occurs through a PKC ζ-dependent mechanism that is independent of vascular endothelial growth factor (VEGF).

  • CO production: HO-1 catalyzes the degradation of heme to produce carbon monoxide (CO), which acts as a second messenger in this pathway.

  • VASP phosphorylation: Vasodilator-stimulated phosphoprotein (VASP), a cytoskeletal-associated protein involved in cell migration, is phosphorylated in response to SDF-1g in an HO-1-dependent manner. This phosphorylation is critical for endothelial cell migration and can be restored by CO in HO-1-deficient cells.

Experimental evidence from in vitro and in vivo models supports this mechanism. For example, aortic rings from HO-1-deficient (HO-1 −/−) mice fail to form capillary sprouts in response to SDF-1g, a defect that can be reversed by CO supplementation. Similarly, endothelial tube formation and migration are impaired in HO-1-deficient cells, and the functional significance of HO-1 in SDF-1g-mediated angiogenesis has been confirmed in Matrigel plug, wound healing, and retinal ischemia models in vivo.

What are the optimal conditions for studying SDF-1g-induced endothelial progenitor cell migration?

For optimal study of SDF-1g-induced endothelial progenitor cell (EPC) migration, researchers should consider:

  • Cell isolation and characterization: EPCs should be isolated and characterized by flow cytometry for expression of CXCR4 and endothelial markers such as CD31. Functional characterization can include DiI-Ac-LDL uptake. Research indicates that approximately 89.3% of properly isolated EPCs express CXCR4 and are positive for CD31.

  • SDF-1g concentration: The effective dose for inducing migration typically ranges from 4-24 ng/mL, with 100 ng/ml being commonly used in research protocols.

  • Migration assay setup: Transwell migration assays with SDF-1g as a chemoattractant in the lower chamber are commonly used. Quantification should include counting the number of migrated cells in multiple high-power fields.

  • Controls: Include both positive controls (known chemoattractants) and negative controls (buffer only). For mechanistic studies, include:

    • EPCs from HO-1 +/+ and HO-1 −/− mice to assess the role of HO-1

    • CXCR4 antagonists (e.g., AMD 3100) to confirm receptor specificity

    • Anti-SDF-1 neutralizing antibodies to demonstrate ligand specificity

  • Time course: Optimal migration is typically observed after 4-6 hours of exposure to SDF-1g.

How can researchers address contradictory findings in SDF-1g studies?

Contradictory findings are common in rapidly evolving fields like SDF-1g research. To address these contradictions, researchers should implement:

  • Systematic literature review: Thoroughly analyze published studies on SDF-1g, noting experimental methods, cell types, concentrations, and endpoints to identify potential sources of variation.

  • Standardized reporting: Follow ARRIVE guidelines for in vivo studies and include detailed methodological descriptions, including:

    • Exact recombinant protein specifications (e.g., E. coli-derived human CXCL12/SDF-1 gamma protein)

    • Cell passage numbers

    • Culture conditions

    • Assay protocols

  • Replication studies: Conduct direct replications of contradictory findings using identical protocols when possible, or systematic variations to identify critical parameters.

  • Multi-parameter analysis: When contradictory findings persist, analyze multiple endpoints simultaneously to develop a more comprehensive understanding of SDF-1g activity.

  • Computational approaches: Leverage distant supervision approaches and clinical ontologies like SNOMED (Systematized Nomenclature of Medicine Clinical Terms) to automatically identify and classify potentially contradictory statements in the literature. This technique can help researchers navigate the large body of sometimes conflicting literature on SDF-1g.

What is the significance of SDF-1 gene polymorphisms in clinical research and how should they be incorporated into study designs?

SDF-1 gene polymorphisms, particularly SDF-1 G801A (rs1801157), have significant clinical implications, especially in hematological conditions. Studies have shown an association between certain SDF-1 polymorphisms and an increased risk of acute myeloid leukemia (AML), with the SDF-1 dominant model conferring a higher risk of developing AML.

When incorporating SDF-1 polymorphism analysis into clinical research designs, researchers should consider:

  • Sample size calculation: For genetic association studies involving SDF-1 polymorphisms, calculate sample size using established statistical methods. For example, a study with a level of absolute precision of 2% at alpha error of 5% and study power of 80% would require at least 54 subjects per group, with 60 subjects recommended to account for potential dropouts.

  • Genotype-phenotype correlations: Design studies to correlate SDF-1 genotypes with:

    • Disease risk or progression

    • Treatment response

    • Receptor expression (e.g., CXCR4 expression levels)

    • Functional outcomes (e.g., cell migration, angiogenesis potential)

  • Complementary receptor analysis: Since SDF-1 functions through receptor interactions, include analysis of CXCR4 expression (with a suggested cutoff of 20.0% in flow cytometry analysis). This is particularly important as CXCR4 positive expression has been found to predict poor prognosis in AML patients and could represent a potential therapeutic target.

  • Translational relevance: Design studies with clear translational goals, such as:

    • Identifying patient subgroups who might benefit from CXCR4 antagonist therapy

    • Developing prognostic or predictive biomarkers based on SDF-1 polymorphisms

    • Understanding the functional consequences of polymorphisms on SDF-1g signaling and angiogenic potential

What are common challenges in producing biologically active recombinant SDF-1g and how can they be addressed?

Researchers frequently encounter challenges when working with recombinant SDF-1g. Here are the common issues and solutions:

  • Protein folding and activity:

    • Challenge: Incorrect folding affecting biological activity.

    • Solution: Use E. coli-derived human CXCL12/SDF-1 gamma protein systems that have demonstrated biological activity, with an ED50 of 4-24 ng/mL in functional assays.

  • N-terminal processing:

    • Challenge: Proteolytic processing of the first two N-terminal amino acids (Lys-Pro) by CD26 can create a reduced-activity chemokine.

    • Solution: Consider using protease inhibitors during preparation or N-terminally modified variants resistant to CD26 processing when studying SDF-1g functions.

  • Heparin binding:

    • Challenge: The highly charged C-terminal domain of SDF-1g with four BBXB motifs has strong heparin binding properties that can affect its distribution and activity.

    • Solution: For in vitro studies, carefully control the presence of heparin/heparan sulfate in the experimental system, as these can significantly influence SDF-1g activity.

  • Receptor specificity:

    • Challenge: Dual binding to CXCR4 and CXCR7 receptors can complicate interpretation of results.

    • Solution: Use selective antagonists (e.g., AMD 3100 for CXCR4) to isolate receptor-specific effects, employing dose-dependent approaches (3-300 nM) to establish specificity.

How can researchers optimize SDF-1g detection in clinical samples?

Optimizing SDF-1g detection in clinical samples requires careful consideration of several factors:

  • Sample collection and processing:

    • Collect blood samples in EDTA tubes (2 mL is typically sufficient for genomic studies)

    • Process samples within 2-4 hours for optimal CXCR4 expression analysis

    • Use standardized protocols for DNA extraction, such as commercial purification kits

  • PCR-RFLP optimization:

    • Use validated primers for SDF-1 gene polymorphism analysis (Forward: 5′ CAG TCA ACC TGG GCA AAG CC 3′; Reverse: 5′ AGC TTT GGT CCT GAG AGT CC 3′)

    • Carefully control PCR cycling conditions, with initial activation at 95°C for 10 minutes

    • Ensure complete digestion with restriction enzymes (10U MspI at 37°C)

  • Flow cytometry for CXCR4 detection:

    • Establish a consistent cutoff (e.g., 20.0%) for CXCR4 positivity

    • Use appropriate fluorochrome-conjugated antibodies

    • Include relevant isotype controls

    • Analyze bone marrow aspirate or venous blood samples within 2-4 hours of collection

  • Validation approaches:

    • Compare results across different detection methods when possible

    • Include known positive and negative controls

    • For clinical studies, calculate appropriate sample sizes based on expected genotype frequencies (e.g., a minimum of 54 subjects per group with alpha error of 5% and study power of 80%)

Product Science Overview

Introduction

Stromal Cell-Derived Factor-1 gamma (SDF-1γ), also known as CXCL12γ, is a member of the CXC chemokine family. Chemokines are small cytokines or signaling proteins secreted by cells, and they play a crucial role in immune responses by directing the migration of circulating leukocytes to sites of inflammation or injury.

Gene and Protein Structure

The CXCL12 gene is located on chromosome 10 in humans and produces several isoforms through alternative splicing . The gamma isoform, SDF-1γ, is a 12 kDa protein that binds heparin and is characterized by a typical chemokine-like fold with three antiparallel beta-strands . The protein is not glycosylated and contains four conserved cysteine residues that form two disulfide bonds, a hallmark of the CXC chemokine family .

Biological Functions

CXCL12γ functions as a ligand for the G-protein coupled receptor CXCR4. This interaction is pivotal in various physiological processes, including:

  • Embryogenesis: Directing the migration of hematopoietic cells from the fetal liver to the bone marrow.
  • Immune Surveillance: Attracting lymphocytes to sites of inflammation.
  • Tissue Homeostasis: Maintaining the microenvironment of various tissues.
  • Tumor Growth and Metastasis: CXCL12 signaling has been observed in several cancers, where it can promote tumor growth and metastasis .
Clinical Significance

The role of CXCL12γ in disease states is significant. It is upregulated in conditions characterized by tissue hypoxia, such as myocardial infarction and ischemic cardiomyopathy . Additionally, the CXCL12 gene contains single nucleotide polymorphisms (SNPs) associated with an increased risk of coronary artery disease .

Recombinant Human CXCL12γ

Recombinant human CXCL12γ is produced in E. coli and is used in various research applications. It is typically purified to a high degree of purity (>95%) and is used to study its biological activities, such as its ability to chemoattract cells expressing CXCR4 . The recombinant protein is available in both carrier-free and carrier-containing formulations, with the latter often containing bovine serum albumin (BSA) to enhance stability .

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.