SDF 1b Rat

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

Description

SDF-1 beta Rat Recombinant produced in E.Coli is a non-glycosylated, Polypeptide chain containing 72 amino acids and having a molecular mass of 8.4 kDa.
The Rat SDF-1b is purified by proprietary chromatographic techniques.

Product Specs

Introduction
SDF-1, or stromal cell-derived factor-1, is a chemokine officially known as Chemokine (C-X-C motif) ligand 12 (CXCL12). It exists in two isoforms, SDF-1α/CXCL12a and SDF-1β/CXCL12b, generated by alternative splicing of the CXCL12 gene. Chemokines are characterized by four conserved cysteines forming two disulfide bonds. CXCL12 proteins belong to the CXC chemokine group, with a single amino acid separating the first cysteine pair. CXCL12 exhibits strong chemotactic activity for lymphocytes and plays a crucial role in cell coordination during development, including directing hematopoietic cell migration from fetal liver to bone marrow during embryogenesis. CXCL12 knockout mice are either lethal before birth or die within an hour after birth. Additionally, CXCL12a influences neuronal electrophysiology. In mice, CXCL12 expression is observed in various tissues, including the brain, thymus, heart, lung, liver, kidney, spleen, and bone marrow. CXCL12 binds to the CXCR4 receptor, previously known as fusin. While this interaction was initially considered exclusive, recent studies suggest that CXCL12 also binds to the CXCR7 receptor. The CXCL12 gene is located on human chromosome 10. Both CXCL12 and CXCR4 exhibit high sequence identity between humans and mice, with 99% and 90% similarity, respectively.
Description
SDF-1 beta Rat Recombinant, produced in E. coli, is a non-glycosylated polypeptide chain composed of 72 amino acids, with a molecular weight of 8.4 kDa. This recombinant protein is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder
Formulation
The Rat SDF1B protein was lyophilized from a concentrated (1mg/ml) sterile solution containing 20mM Phosphate buffer at pH 7.4 and 0.15M NaCl.
Solubility
To reconstitute the lyophilized SDF-1b, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions.
Stability
Lyophilized SDF-1b remains stable at room temperature for up to 3 weeks; however, it should be stored desiccated at a temperature below -18°C. Upon reconstitution, CXCL12 should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to store it at a temperature below -18°C. To enhance long-term storage stability, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined by the following methods and is greater than 97.0%: (a) Analysis by RP-HPLC (b) Analysis by SDS-PAGE
Biological Activity
The specific activity of this protein is determined by its chemoattractant properties. It can effectively chemoattract human peripheral blood monocytes at concentrations between 100-150 ng/ml, corresponding to a specific activity ranging from 6,667 to 10,000 IU/mg.
Synonyms
SDF-1, CXCL12, Pre-B cell growth-stimulating factor, PBSF, hIRH, chemokine (C-X-C motif) ligand 12, SDF1, SDF1B, TPAR1, SCYB12, SDF-1b, TLSF-b, 12-O-tetradecanoylphorbol 13-acetate repressed protein 1, Thymic lymphoma cell-stimulating factor, TLSF.
Source
Escherichia Coli.
Amino Acid Sequence
KPVSLSYRCP CRFFESHVAR ANVKHLKILN TPNCALQIVA RLKSNNRQVC IDPKLKWIQE YLDKALNKRL KM.

Q&A

What is SDF-1β and how does it differ from other SDF-1 isoforms in rats?

SDF-1β is one of the isoforms of Stromal cell-derived factor 1 (also known as CXCL12), a chemokine that plays crucial roles in various biological processes including tissue repair, stem cell migration, and organ development. In rats, as in other mammals, SDF-1 exists in multiple isoforms, with SDF-1α and SDF-1β being the most studied. These isoforms differ in their C-terminal sequences, with SDF-1β containing a 4-amino acid extension compared to SDF-1α . This structural difference contributes to variations in their binding affinity to receptors and subsequent biological activities. Both isoforms signal primarily through the CXCR4 receptor, stimulating cellular signals that attract stem cells to injury sites for tissue repair and remodeling .

What are the key signaling pathways associated with SDF-1β in rat models?

SDF-1β primarily signals through the SDF-1/CXCR4 signaling pathway in rats. This pathway is critical for:

  • Mobilization and recruitment of bone marrow mesenchymal stem cells (BMSCs) to injury sites

  • Promotion of cell migration, proliferation, and differentiation

  • Regulation of apoptosis in specific cell populations

  • Modulation of inflammatory responses

The expression of SDF-1 is regulated by hypoxia-inducible factor 1α (HIF-1α), which increases under hypoxic conditions such as those created during tissue distraction. This leads to enhanced SDF-1 expression, which then recruits CXCR4-positive progenitor cells (including endothelial progenitor cells and BMSCs) to promote tissue regeneration . The pathway can be experimentally inhibited using compounds such as AMD3100, which blocks the interaction between SDF-1 and CXCR4, allowing researchers to study the specific contributions of this signaling axis to various biological processes .

What physiological processes in rats are influenced by SDF-1β?

Based on current research, SDF-1β in rats influences several key physiological processes:

  • Tissue repair and regeneration: SDF-1β plays a crucial role in attracting stem cells to sites of injury for tissue repair .

  • Bone formation and remodeling: It promotes bone regeneration and mineralization during distraction osteogenesis by recruiting bone marrow mesenchymal stem cells .

  • Pulmonary function and fibrosis: SDF-1β overexpression can attenuate lung fibrosis in bleomycin-injured rat lungs by inducing myofibroblast apoptosis and promoting alveolar epithelial cell proliferation .

  • Cardiovascular function: The SDF-1/CXCR4 axis shows altered expression in the ventricles of rats with pulmonary hypertension, suggesting its involvement in cardiac adaptation to pressure overload .

  • Cell migration and chemotaxis: SDF-1β serves as a potent chemoattractant for various cell types, directing their migration to specific locations .

What are the established protocols for inducing overexpression of SDF-1β in rat lung models?

Overexpression of SDF-1β in rat lung models can be achieved through in vivo electroporation-mediated gene transfer, as demonstrated in studies of bleomycin-induced lung injury. The procedure typically follows this methodology:

  • Preparation of animal model: Intratracheal administration of bleomycin (or other fibrosis-inducing agent) is performed in adult male rats (such as F344 strain).

  • Timing of intervention: Seven days after bleomycin administration, when initial fibrotic changes have been established but before irreversible fibrosis occurs.

  • Gene transfer procedure:

    • Plasmid preparation: SDF-1β cDNA is cloned into an appropriate expression vector.

    • Anesthesia: Rats are anesthetized using established protocols for the procedure.

    • Delivery method: Direct intratracheal instillation of plasmid DNA.

    • Electroporation: Application of electrical pulses using specialized electrodes positioned on the chest wall to enhance DNA uptake by cells in the lung.

  • Control groups: Include rats receiving the empty vector through the same delivery method.

  • Evaluation timeline: Tissue collection and analysis are typically performed seven days after gene transfer to assess the effects of SDF-1β overexpression .

This approach allows for targeted gene transfer to the lungs while minimizing systemic effects and has been shown to successfully induce SDF-1β overexpression in rat lung tissue.

How can researchers effectively measure SDF-1β expression levels in rat tissue samples?

Accurate measurement of SDF-1β expression in rat tissue samples can be achieved through several complementary techniques:

Protein-level detection:

  • Western blotting:

    • Tissue homogenization in appropriate lysis buffer

    • Protein quantification using Bradford method with bovine serum albumin as standard

    • SDS-PAGE gel electrophoresis for protein separation

    • Transfer to nitrocellulose membrane

    • Blocking and incubation with primary antibodies (e.g., rabbit polyclonal to SDF-1, AB 25117, 1:1,000; Abcam)

    • Incubation with appropriate secondary antibodies (e.g., horseradish peroxidase-conjugated anti-rabbit IgG, AB 6721, 1:2,000; Abcam)

    • Visualization using enhanced chemiluminescence and quantification with software such as TotalLab

  • Enzyme-linked immunosorbent assay (ELISA): Commercial kits specific for rat SDF-1β are available for quantitative measurement in tissue homogenates or biological fluids.

mRNA-level detection:

  • Quantitative real-time PCR (qRT-PCR):

    • RNA extraction from tissue samples

    • cDNA synthesis through reverse transcription

    • Amplification using primers specific for rat SDF-1β

    • Normalization to appropriate housekeeping genes

    • Relative quantification using the 2^(-ΔΔCT) method

Localization studies:

  • Immunohistochemistry or immunofluorescence to visualize the spatial distribution of SDF-1β within tissue sections

  • In situ hybridization to detect SDF-1β mRNA expression at the cellular level

For optimal results, researchers should consider using multiple detection methods to confirm findings and include appropriate positive and negative controls to validate the specificity of the assays.

What is the recommended method for delivering exogenous SDF-1β to rat models in distraction osteogenesis studies?

Based on successful research in distraction osteogenesis (DO) models, the recommended method for delivering exogenous SDF-1β to rat models involves:

Preparation and timing:

  • Establish the DO model through appropriate surgical procedures (e.g., mid-femoral osteotomy fixed with an external fixator).

  • Allow for a latency period (typically 5 days) before beginning distraction.

  • Perform distraction at a controlled rate (e.g., 0.25 mm every 12 hours) for the desired duration (typically 10 days for a total lengthening of 5 mm).

  • Begin SDF-1β administration after completing the distraction phase.

Delivery method:

  • Direct local injection into the osteotomy site is the most effective approach.

  • Recommended dosage: 200 ng of SDF-1β in 1 ml of PBS solution.

  • Administration frequency: Once daily for 7 consecutive days.

  • For studies examining synergistic effects, SDF-1β can be combined with other growth factors (such as VEGF at 50 ng) in the same injection solution.

Control groups:

  • Vehicle control: PBS injections of equal volume.

  • Comparison groups: Single growth factor injections when studying combinatorial effects.

Evaluation methods:

  • Sequential radiographic monitoring (recommended every 2 weeks).

  • Micro-CT analysis of bone density and volume.

  • Biomechanical testing of regenerate strength.

  • Histological examination of new bone formation.

  • Molecular analysis of osteogenic gene expression .

This protocol has been demonstrated to significantly enhance bone regeneration and mineralization during DO in rat models, with measurable improvements in both radiographic and histological outcomes.

How does SDF-1β overexpression specifically affect myofibroblast populations in bleomycin-injured rat lungs?

SDF-1β overexpression has been shown to have significant and specific effects on myofibroblast populations in bleomycin-injured rat lungs, primarily through the induction of apoptosis. Research demonstrates the following mechanistic actions:

  • Selective induction of myofibroblast apoptosis: Seven days after SDF-1β gene transfer to bleomycin-injured rat lungs, significant myofibroblast apoptosis was observed. This effect appears to be specific to activated myofibroblasts, which are the key cellular mediators of fibrosis .

  • TNF-α-mediated apoptotic pathway: Molecular studies have revealed that SDF-1β-induced apoptosis of myofibroblasts is mediated by TNF-α. This suggests that SDF-1β activates signaling cascades that increase local TNF-α production or sensitivity, which then triggers apoptotic pathways in myofibroblasts .

  • Reduced extracellular matrix deposition: As a consequence of myofibroblast apoptosis, there is decreased production and deposition of extracellular matrix components, particularly collagen. This is evidenced by:

    • Reduced total collagen content in lung tissue

    • Decreased collagen fibril density in the alveolar interstitium

    • Improved histological parameters of fibrosis

  • Cell-specific effects: Importantly, while SDF-1β promotes apoptosis in myofibroblasts, it simultaneously increases alveolar epithelial cell numbers and stimulates their proliferation in vivo. This dual action—removing the fibrosis-promoting cells while supporting the regeneration of functional epithelium—creates a coordinated anti-fibrotic effect that contributes to improved lung structure and potentially function .

These findings highlight the potential of SDF-1β as a therapeutic approach that selectively targets activated myofibroblasts while promoting beneficial epithelial responses, addressing a fundamental challenge in developing treatments for pulmonary fibrosis.

What are the synergistic mechanisms between SDF-1 and VEGF in bone regeneration in rat models?

Research using rat distraction osteogenesis models has revealed several synergistic mechanisms between SDF-1 and VEGF that enhance bone regeneration:

  • Complementary cellular recruitment: SDF-1 primarily recruits bone marrow mesenchymal stem cells (BMSCs) through the SDF-1/CXCR4 signaling pathway, while VEGF attracts endothelial progenitor cells (EPCs). Together, they create an optimal cellular environment for coordinated bone formation and vascularization .

  • Enhanced angiogenesis: VEGF is a potent angiogenic factor that stimulates new blood vessel formation, while SDF-1 supports this process by recruiting EPCs and promoting their differentiation into endothelial cells. The increased vascularity provides essential nutrients and oxygen to the regenerating bone tissue, creating a supportive microenvironment for osteogenesis .

  • Amplified osteogenic gene expression: Combined administration of SDF-1 and VEGF results in significantly higher expression of osteogenic-specific genes compared to either factor alone. This suggests a molecular cross-talk that amplifies osteogenic differentiation signals .

  • Accelerated mineralization: Radiographic and micro-CT analyses demonstrate that the combination of SDF-1 and VEGF leads to more rapid bridging of the distraction gap and higher bone mineral density than single-factor treatments. By week 4 after treatment, the SDF-1+VEGF group showed nearly complete bridging of the gap with higher-density callus formation .

  • Superior mechanical properties: Biomechanical testing reveals that regenerate bone in the SDF-1+VEGF group has significantly higher mechanical maximum loading capacity, indicating that the newly formed bone not only appears more abundant radiographically but is also functionally superior .

  • Improved histological maturation: Histological examination shows more abundant new trabeculae in the distraction gap and more mature bone tissue architecture in the combined treatment group, suggesting that the synergistic effect influences not only the quantity but also the quality of new bone formation .

These mechanisms highlight the potential of combination therapy with SDF-1 and VEGF for enhancing bone regeneration in clinical applications such as fracture healing, bone defect repair, and distraction osteogenesis.

How does the expression pattern of SDF-1 and its receptor CXCR4 change in rat cardiac ventricles during pulmonary hypertension progression?

The expression pattern of SDF-1 and its receptor CXCR4 undergoes dynamic changes in rat cardiac ventricles during the progression of pulmonary hypertension, as demonstrated in the monocrotaline-induced model:

These findings suggest that the SDF-1/CXCR4 axis plays different roles in the adaptation of each ventricle to pulmonary hypertension, potentially influencing processes such as myocardial remodeling, inflammation, and compensation mechanisms in response to altered hemodynamics.

What control experiments are essential when studying SDF-1β effects in rat models of lung fibrosis?

When designing experiments to study SDF-1β effects in rat models of lung fibrosis, the following control experiments are essential to ensure valid and interpretable results:

  • Vehicle controls:

    • Empty vector control: Rats receiving the same gene delivery method but with an empty vector (no SDF-1β cDNA)

    • Vehicle-only control: Rats receiving the delivery vehicle without plasmid

    • Sham procedure control: Rats undergoing the delivery procedure without actual gene transfer

  • Disease model controls:

    • Healthy control: Rats receiving no bleomycin and no treatment

    • Disease model only: Rats receiving bleomycin but no intervention

    • Timing controls: Separate groups sacrificed at different time points to establish the natural progression of fibrosis

  • Dose-response controls:

    • Multiple groups receiving different doses of SDF-1β to establish dose-dependent effects

    • Time-course experiments with fixed doses administered at different time points relative to bleomycin injury

  • Pathway validation controls:

    • SDF-1 receptor blockade: Co-administration of SDF-1β with CXCR4 antagonists (e.g., AMD3100)

    • TNF-α pathway inhibition: Since TNF-α mediates SDF-1β-induced myofibroblast apoptosis, including TNF-α inhibitors would confirm this mechanism

    • Apoptosis inhibition: Administration of pan-caspase inhibitors to confirm the role of apoptosis in SDF-1β effects

  • Cell-specific controls:

    • Myofibroblast-specific markers: α-SMA staining to quantify myofibroblast populations

    • Epithelial cell markers: Specific staining for alveolar epithelial cells to confirm proliferation effects

    • Co-localization studies: Dual staining for cell type markers and apoptosis markers to confirm cell-specific effects

  • Alternative anti-fibrotic agents:

    • Positive control: Administration of known anti-fibrotic agents (e.g., pirfenidone or nintedanib)

    • Combination therapy: SDF-1β plus established anti-fibrotics to assess additive or synergistic effects

These comprehensive controls ensure that observed effects can be specifically attributed to SDF-1β, establish dose-response relationships, validate proposed mechanisms, and provide comparative data against established treatments.

How can researchers accurately quantify the anti-fibrotic effects of SDF-1β in rat lung tissue?

Accurate quantification of the anti-fibrotic effects of SDF-1β in rat lung tissue requires a multi-modal approach that combines histological, biochemical, molecular, and functional assessments:

  • Histological quantification:

    • Masson's trichrome staining: For visualization and quantification of collagen deposition

    • Picrosirius red staining: For specific detection of fibrillar collagens with enhanced contrast

    • Design-based stereology: Utilizes systematic random sampling to provide unbiased quantitative data on:

      • Volume fraction of fibrotic tissue

      • Total volume of fibrotic tissue

      • Septal wall thickness

      • Alveolar surface area

    • Immunohistochemistry for fibrosis markers: α-SMA (for myofibroblasts), fibronectin, and various collagen subtypes

    • Ashcroft scoring: Semi-quantitative assessment of fibrosis severity on a scale of 0-8

  • Biochemical quantification:

    • Hydroxyproline assay: Quantifies total collagen content in lung tissue

    • Sircol collagen assay: Colorimetric method for measuring soluble collagens

    • Collagen fibril measurement: Quantification of individual collagen fibrils using electron microscopy

  • Molecular quantification:

    • qRT-PCR for fibrosis-related genes: Col1a1, Col3a1, Fn1, α-SMA, TGF-β1, TIMP

    • Western blot analysis: Quantification of fibrosis-related proteins

    • Multiplex cytokine assays: Measurement of pro-fibrotic and anti-fibrotic mediators in lung tissue homogenates

  • Cellular quantification:

    • Flow cytometry: For quantification of myofibroblasts, inflammatory cells, and apoptotic cells

    • TUNEL assay: For detection and quantification of apoptotic cells

    • BrdU or Ki67 staining: For quantification of proliferating cells, particularly alveolar epithelial cells

  • Functional assessment:

    • Pulmonary function tests: Measurement of lung compliance, resistance, and vital capacity

    • Oxygen saturation: Non-invasive measure of lung function

    • Exercise capacity: Indirect measurement of pulmonary function improvement

  • Computational analysis:

    • Digital image analysis: Automated quantification of histological parameters

    • Machine learning algorithms: For unbiased classification of fibrotic changes

    • 3D reconstruction: To assess the spatial distribution of fibrotic lesions

By combining these complementary approaches, researchers can obtain a comprehensive assessment of the anti-fibrotic effects of SDF-1β, encompassing structural, biochemical, cellular, and functional improvements in rat lung tissue.

What statistical approaches are most appropriate for analyzing the synergistic effects of SDF-1 and VEGF in bone regeneration studies?

When analyzing the synergistic effects of SDF-1 and VEGF in bone regeneration studies, several specialized statistical approaches are recommended to properly characterize interaction effects and account for the complex, multidimensional nature of the data:

  • Factorial analysis for synergy detection:

    • Two-way ANOVA with interaction term: Essential for determining whether the combined effect of SDF-1 and VEGF is greater than the sum of their individual effects. The interaction term specifically tests for synergy.

    • Post-hoc testing: Tukey's HSD or Bonferroni correction for multiple comparisons between treatment groups (control, SDF-1 alone, VEGF alone, SDF-1+VEGF) .

  • Specialized synergy metrics:

    • Synergy factor calculation: SF = (Combined effect)/(Effect of A + Effect of B - Effect of A × Effect of B)

    • Loewe additivity model: Based on dose-effect curves to distinguish between additive and synergistic effects

    • Bliss independence model: Evaluates whether combined effects exceed the predicted independent actions

  • Longitudinal data analysis:

    • Repeated measures ANOVA: For analyzing data collected across multiple time points (e.g., sequential X-rays at 2, 4, 6, and 8 weeks)

    • Mixed-effects models: To account for both fixed effects (treatments) and random effects (individual differences between animals)

    • Growth curve modeling: To characterize the rate and pattern of bone regeneration over time

  • Multivariate approaches for integrated analysis:

    • MANOVA: For simultaneous analysis of multiple dependent variables (e.g., bone mineral density, bone volume, mechanical loading capacity)

    • Principal component analysis: To reduce dimensionality and identify patterns across multiple outcome measures

    • Canonical correlation analysis: To examine relationships between sets of variables (e.g., imaging parameters and biomechanical properties)

  • Non-parametric alternatives:

    • Kruskal-Wallis test with Dunn's post-hoc test: When data do not meet assumptions of normality

    • Friedman test: Non-parametric alternative for repeated measures designs

    • Permutation tests: For robust inference without distributional assumptions

  • Sample size and power considerations:

    • A priori power analysis: Specifically designed to detect interaction effects, which typically require larger sample sizes than main effects

    • Adjustment for multiple endpoints: Controlling family-wise error rate or false discovery rate when analyzing multiple outcome measures

    • Sequential design considerations: For studies with interim analyses at predefined time points

When reporting results, researchers should clearly specify:

  • The statistical models used to test for synergy

  • The specific definition of synergy being applied

  • Effect sizes and confidence intervals, not just p-values

  • Graphical representations that clearly visualize interaction effects

These approaches allow for rigorous statistical assessment of synergistic effects between SDF-1 and VEGF in bone regeneration, distinguishing true biological synergy from simple additive effects.

What are the key challenges in maintaining consistent SDF-1β expression levels in rat models, and how can they be addressed?

Maintaining consistent SDF-1β expression levels in rat models presents several technical challenges that can impact experimental reproducibility and interpretation. Here are the key challenges and recommended solutions:

  • Challenge: Variability in gene transfer efficiency

    • Solution:

      • Optimize electroporation parameters (voltage, pulse duration, number of pulses) specifically for the target tissue

      • Use reporter genes (e.g., GFP) in pilot studies to visualize and quantify transfection efficiency

      • Employ viral vectors (adeno-associated virus or lentivirus) as alternatives to electroporation for more consistent gene delivery

      • Standardize plasmid preparation protocols to ensure consistent DNA quality and concentration

  • Challenge: Temporal dynamics of expression

    • Solution:

      • Perform time-course studies to characterize the expression profile after gene transfer

      • Consider using inducible expression systems (e.g., tetracycline-inducible) for controlled timing of expression

      • Implement serial sampling methods (e.g., bronchoalveolar lavage) to monitor expression over time in the same animal

      • Design constructs with appropriate promoters that maintain stable expression over the desired timeframe

  • Challenge: Individual variability between animals

    • Solution:

      • Increase sample sizes to account for biological variability

      • Use inbred rat strains to reduce genetic variability

      • Implement strict inclusion/exclusion criteria based on baseline parameters

      • Employ paired experimental designs where possible (e.g., comparing different lung lobes within the same animal)

      • Confirm SDF-1β expression levels in each animal and stratify analyses accordingly

  • Challenge: Protein stability and bioavailability

    • Solution:

      • For exogenous SDF-1β administration, use stabilized formulations (e.g., PEGylated forms)

      • Implement controlled-release delivery systems (e.g., biodegradable microspheres)

      • Consider fusion proteins that enhance half-life while maintaining bioactivity

      • Administer at appropriate frequency based on pharmacokinetic studies

  • Challenge: Neutralizing antibodies or receptor downregulation

    • Solution:

      • Monitor for development of anti-SDF-1β antibodies in long-term studies

      • Assess CXCR4 receptor expression levels throughout the experiment

      • Use SDF-1β analogs that retain signaling capacity but have reduced immunogenicity

      • Consider co-administration of agents that prevent receptor downregulation

  • Challenge: Tissue-specific regulation and feedback mechanisms

    • Solution:

      • Characterize the endogenous regulation of SDF-1β in the target tissue

      • Consider the use of tissue-specific promoters for gene transfer

      • Account for potential feedback inhibition in experimental design

      • Monitor expression of related chemokines that might compensate for altered SDF-1β levels

How can researchers overcome the challenge of distinguishing between direct and indirect effects of SDF-1β in complex in vivo systems?

Distinguishing between direct and indirect effects of SDF-1β in complex in vivo systems presents a significant challenge in experimental biology. The following methodological approaches can help researchers address this challenge:

  • Cell-specific genetic manipulation approaches:

    • Conditional knockout/knockin models: Generate rats with cell-type-specific deletion or overexpression of CXCR4 (the SDF-1β receptor) using Cre-loxP systems

    • Cell-specific promoters: Drive SDF-1β expression under the control of cell-type-specific promoters to localize expression to particular cell populations

    • Inducible systems: Use temporal control of gene expression (e.g., tetracycline-inducible systems) to distinguish between immediate direct effects and delayed indirect effects

  • Receptor antagonism strategies:

    • Selective CXCR4 antagonists: Administer AMD3100 or other specific CXCR4 antagonists to block direct SDF-1β signaling

    • Cell-specific receptor blockade: Deliver CXCR4 antagonists to specific cell populations using targeted nanoparticles

    • Dose-response studies: Compare partial vs. complete receptor blockade to identify threshold effects

  • Ex vivo and in vitro validation:

    • Primary cell isolation: Extract specific cell populations (e.g., myofibroblasts, epithelial cells) from treated animals and analyze them ex vivo

    • Co-culture systems: Recreate cell-cell interactions in controllable in vitro settings to dissect intercellular signaling

    • Conditioned media experiments: Use media from SDF-1β-treated cells to stimulate naïve cells and identify secreted mediators of indirect effects

  • Molecular pathway dissection:

    • Temporal profiling: Perform time-course experiments to establish the sequence of molecular events

    • Pathway-specific inhibitors: Selectively block downstream mediators (e.g., TNF-α) to interrupt specific signaling cascades

    • Phospho-proteomics: Identify immediate signaling events triggered by SDF-1β binding to CXCR4

  • Advanced imaging approaches:

    • Intravital microscopy: Directly visualize cell behavior in living animals following SDF-1β administration

    • Reporter systems: Use fluorescent or bioluminescent reporters linked to pathway activation

    • Spatial transcriptomics: Map gene expression changes with spatial resolution to identify localized vs. spreading effects

  • Multi-omics integration:

    • Single-cell RNA sequencing: Identify cell-specific responses to SDF-1β across all cell types in the tissue

    • Spatial proteomics: Map protein changes with spatial resolution to distinguish local from distant effects

    • Network analysis: Apply computational approaches to distinguish direct signaling nodes from secondary response networks

  • Pharmacological validation:

    • Specific mediator blockade: Identify and block candidate mediators of indirect effects (e.g., TNF-α for myofibroblast apoptosis)

    • Timing of interventions: Apply blockers at different time points to distinguish immediate from delayed effects

    • Ex vivo tissue slices: Maintain tissue architecture while allowing controlled drug application and pathway blocking

What are the best practices for optimizing SDF-1 and VEGF combination therapy in distraction osteogenesis rat models?

Optimizing SDF-1 and VEGF combination therapy in distraction osteogenesis (DO) rat models requires careful consideration of multiple experimental parameters. The following best practices are recommended based on current research:

  • Optimal timing of administration:

    • Initiate treatment immediately after distraction completion: Research demonstrates that beginning treatment at this phase capitalizes on the hypoxic environment created by distraction, which enhances cellular responsiveness to both factors .

    • Duration of treatment: A 7-day daily administration protocol has shown significant efficacy, though extended treatment periods should be investigated for potentially enhanced outcomes .

    • Consider sequential administration: Testing whether sequential delivery (e.g., VEGF followed by SDF-1) might further optimize the regenerative response by first establishing vascularization before stem cell recruitment.

  • Dosage optimization:

    • Established effective doses: 200 ng of SDF-1 combined with 50 ng of VEGF in 1 ml of PBS has demonstrated significant synergistic effects .

    • Dose-ratio exploration: Systematic testing of different SDF-1:VEGF ratios to identify optimal synergistic combinations.

    • Dose-response studies: Establishing full dose-response curves for the combination to identify potential ceiling effects or optimal therapeutic windows.

  • Delivery system refinement:

    • Controlled-release systems: Incorporate SDF-1 and VEGF into biodegradable microspheres, hydrogels, or scaffolds for sustained release.

    • Biomaterial carriers: Design carriers that protect the factors from degradation while maintaining their bioactivity.

    • Spatially controlled delivery: Develop systems that release the factors with defined spatial gradients to direct cell migration and tissue formation.

  • Animal model considerations:

    • Standardize surgical technique: Ensure consistent osteotomy location, gap size, and fixation stability.

    • Control distraction protocol: Maintain consistent distraction rate (0.25 mm/12h) and total lengthening (5 mm) .

    • Animal age and sex: Use age-matched animals and consider including both sexes to assess potential sex-dependent responses.

    • Pre-operative baseline measurements: Establish baseline parameters for each animal to enable paired statistical analyses.

  • Comprehensive outcome assessment:

    • Multi-modal imaging: Combine sequential X-rays with micro-CT analysis at defined intervals (2, 4, 6, and 8 weeks post-distraction) .

    • Quantitative histomorphometry: Assess new bone formation, vascularization, and cellular composition using standardized methods.

    • Biomechanical testing: Perform standardized mechanical testing to assess functional outcomes.

    • Molecular analyses: Quantify expression of osteogenic, angiogenic, and stem cell markers to elucidate mechanisms.

  • Mechanistic investigation:

    • Pathway inhibition studies: Selectively block SDF-1/CXCR4 or VEGF signaling to demonstrate specific contributions.

    • Cell tracking experiments: Label and track BMSCs and endothelial progenitor cells to visualize recruitment and differentiation.

    • Temporal analysis: Collect samples at multiple time points to establish the sequence of cellular and molecular events.

  • Translation-focused approaches:

    • Clinically relevant delivery methods: Prioritize delivery systems that could be readily translated to clinical applications.

    • Comparative studies: Compare SDF-1/VEGF combination with current clinical standards of care.

    • Functional recovery assessment: Include functional measures relevant to clinical outcomes, not just tissue regeneration markers.

Implementing these best practices will help researchers optimize SDF-1 and VEGF combination therapy for enhanced bone regeneration in distraction osteogenesis, potentially reducing consolidation time and improving functional outcomes in clinical applications.

What are the most promising translational applications of SDF-1β research in rat models for human disease treatment?

Based on current research in rat models, several translational applications of SDF-1β show particular promise for human disease treatment:

Translation of these applications from rat models to human treatments will require addressing several challenges, including optimizing delivery methods, ensuring sustained therapeutic levels, managing potential off-target effects, and developing clinically feasible administration protocols. Nevertheless, the mechanistic insights and therapeutic effects demonstrated in rat models provide a strong foundation for advancing SDF-1β-based approaches toward clinical applications.

What novel delivery systems for SDF-1β should be explored to enhance its efficacy in rat models?

Several innovative delivery systems for SDF-1β warrant exploration to enhance its efficacy, bioavailability, and targeting specificity in rat models:

  • Advanced biomaterial-based delivery systems:

    • Injectable hydrogels: Thermosensitive or pH-responsive hydrogels that solidify in situ, providing controlled release of SDF-1β at the target site. These systems are particularly suitable for applications like bone regeneration or localized anti-fibrotic therapy .

    • Nanofiber scaffolds: Electrospun nanofibers that mimic extracellular matrix architecture while providing sustained SDF-1β release, creating both structural support and biochemical signals.

    • 3D-printed constructs: Patient-specific implants with defined architecture and embedded SDF-1β-releasing compartments, especially valuable for complex bone defects.

    • Self-assembling peptide hydrogels: Biocompatible matrices that form nanofibrous networks capable of binding SDF-1β and controlling its release through enzymatic degradation.

  • Nanoparticle-based delivery approaches:

    • PLGA nanoparticles: Biodegradable polymer particles that can protect SDF-1β from degradation while allowing controlled release over weeks to months.

    • Liposomal formulations: Lipid vesicles that enhance SDF-1β stability and cellular uptake, potentially enabling systemic administration with reduced proteolytic degradation.

    • Exosome-mimetic nanoparticles: Engineered vesicles that mimic natural extracellular vesicles, potentially enhancing bioactivity and cellular targeting.

    • Inhalable nanoparticles: For pulmonary applications, specially formulated particles that can deliver SDF-1β directly to the lungs through inhalation, maximizing local concentration while minimizing systemic exposure .

  • Gene therapy approaches:

    • Improved viral vectors: Next-generation adeno-associated viral (AAV) vectors with enhanced tropism for specific target tissues and reduced immunogenicity.

    • Non-viral gene delivery systems: Lipid nanoparticles, polymer complexes, or physical methods (improved electroporation) that offer safer alternatives to viral vectors .

    • mRNA delivery platforms: Systems that deliver mRNA encoding SDF-1β for transient expression, potentially offering better safety profiles than DNA-based approaches.

    • Genome editing technologies: CRISPR-Cas9 or base editing approaches to enhance endogenous SDF-1β expression in specific cell populations.

  • Responsive and intelligent delivery systems:

    • Stimuli-responsive release systems: Materials that release SDF-1β in response to specific disease-associated signals (e.g., matrix metalloproteinases in fibrosis or inflammatory cytokines).

    • Magnetically guided delivery: Magnetic nanoparticles conjugated with SDF-1β that can be directed to specific sites using external magnetic fields.

    • Ultrasound-triggered release: Acoustically responsive particles that release SDF-1β upon exposure to focused ultrasound, enabling precise spatial and temporal control.

    • Cell-mediated delivery: Engineered cells (e.g., mesenchymal stem cells) that serve as production factories for continuous SDF-1β secretion at target sites.

  • Combination delivery platforms:

    • Dual-factor delivery systems: Platforms specifically designed to co-deliver SDF-1 and VEGF with optimized release kinetics for each factor, enhancing their synergistic effects .

    • Sequential release systems: Materials engineered to release multiple factors in a predetermined sequence (e.g., VEGF followed by SDF-1β) to mimic natural regenerative processes.

    • Cell-factor combination platforms: Scaffolds that simultaneously deliver SDF-1β and support cells (e.g., stem cells or progenitor cells) for enhanced regenerative outcomes.

  • Targeted delivery approaches:

    • Antibody-conjugated carriers: SDF-1β conjugated to antibodies that recognize specific cell types or tissue components to enhance targeting.

    • Aptamer-based targeting: Nucleic acid aptamers that bind to specific cell surface markers for targeted delivery.

    • Cell-penetrating peptides: Short peptide sequences that enhance cellular uptake of SDF-1β or its carriers.

Exploring these novel delivery systems in rat models could significantly enhance the translational potential of SDF-1β-based therapies by addressing key challenges related to protein stability, controlled release, targeted delivery, and sustained bioactivity.

What emerging research questions remain unanswered regarding the role of SDF-1β in rat models of disease and regeneration?

Despite significant advances in understanding SDF-1β's functions in rat models, several critical research questions remain unexplored or insufficiently addressed:

  • Isoform-specific functions and regulation:

    • How do the biological functions of SDF-1β differ from those of SDF-1α and other isoforms in various rat tissues and disease models?

    • What are the specific transcriptional and post-transcriptional mechanisms regulating SDF-1β expression in different rat tissues during health and disease?

    • Are there tissue-specific or disease-specific variations in SDF-1β processing or secretion that influence its biological activity?

  • Cellular mechanisms and signaling pathways:

    • What are the complete signaling networks through which SDF-1β induces myofibroblast apoptosis while promoting epithelial cell proliferation in lung fibrosis models?

    • How does TNF-α specifically mediate SDF-1β-induced apoptosis of myofibroblasts, and what determines cellular susceptibility to this effect?

    • Beyond CXCR4, what role do alternative receptors (e.g., CXCR7/ACKR3) play in mediating SDF-1β effects in rat models?

  • Temporal dynamics and long-term effects:

    • What are the long-term effects of SDF-1β overexpression or administration in rat models beyond the timeframes currently studied?

    • How does repeated or sustained SDF-1β exposure affect receptor expression, signaling sensitivity, and biological responses?

    • What is the optimal timing for SDF-1β intervention in different disease models to maximize therapeutic effects?

  • Integration with other biological systems:

    • How does SDF-1β interact with the immune system in rat models, particularly regarding inflammation resolution and tissue repair?

    • What is the relationship between SDF-1β signaling and other major regenerative or fibrotic pathways (e.g., TGF-β, Wnt, Hedgehog signaling)?

    • How do age, sex, and comorbidities influence SDF-1β expression and responsiveness in rat models?

  • Disease-specific mechanisms:

    • Beyond pulmonary fibrosis and bone regeneration, what are the effects of SDF-1β modulation in rat models of other fibrotic diseases, such as liver cirrhosis, renal fibrosis, or cardiac fibrosis?

    • How does the SDF-1/CXCR4 axis function in rat models of neurodegenerative diseases, and could SDF-1β have neuroprotective or neuroregenerative effects?

    • What role does SDF-1β play in rat models of metabolic diseases such as diabetes or non-alcoholic steatohepatitis?

  • Translational considerations:

    • How do the effects of SDF-1β in rat models compare with those in larger animal models that more closely approximate human physiology?

    • What are the potential off-target effects or safety concerns associated with long-term SDF-1β overexpression or administration?

    • How can SDF-1β delivery systems be optimized for specific disease applications to maximize efficacy while minimizing side effects?

  • Genetic and epigenetic regulation:

    • What are the epigenetic mechanisms controlling SDF-1β expression in different rat tissues during development, homeostasis, and disease?

    • How do genetic polymorphisms in the SDF-1 gene or its regulatory elements affect expression levels and responses to injury in different rat strains?

    • Can gene editing approaches targeting SDF-1β expression be developed as potential therapeutic strategies for various diseases?

  • Stem cell biology and regenerative potential:

    • What is the full spectrum of stem and progenitor cell populations responsive to SDF-1β in different rat tissues?

    • How does SDF-1β influence stem cell fate decisions beyond migration, including self-renewal, differentiation, and senescence?

    • Can SDF-1β be used to enhance endogenous regenerative processes in tissues with limited regenerative capacity, such as the central nervous system or cardiac tissue?

Addressing these emerging questions will require innovative experimental approaches, including advanced imaging techniques, single-cell analyses, computational modeling, and integrated multi-omics studies to fully elucidate the complex biology of SDF-1β in rat models of disease and regeneration.

What are the key takeaways from current research on SDF-1β in rat models?

Current research on SDF-1β in rat models has yielded several significant insights that collectively advance our understanding of its biological roles and therapeutic potential:

  • Anti-fibrotic mechanisms: SDF-1β overexpression demonstrates remarkable anti-fibrotic effects in bleomycin-injured rat lungs through a dual mechanism: inducing apoptosis of myofibroblasts (mediated by TNF-α) while simultaneously promoting alveolar epithelial cell proliferation and migration. This selective cellular effect makes it a promising candidate for treating pulmonary fibrosis .

  • Synergistic regenerative potential: When combined with VEGF, SDF-1 demonstrates powerful synergistic effects in promoting bone regeneration and mineralization in distraction osteogenesis models. This combination accelerates bone formation, increases bone mineral density, enhances mechanical properties, and improves histological maturation of newly formed bone .

  • Dynamic expression in disease states: The SDF-1/CXCR4 axis shows tissue-specific and temporal regulation in response to pathological conditions, as evidenced by the differential expression patterns observed in right and left ventricles during the progression of pulmonary hypertension. This suggests context-dependent roles in disease processes .

  • Critical role in stem cell homing: SDF-1 functions as a powerful chemotactic factor that recruits bone marrow mesenchymal stem cells and other progenitor cells to sites of injury through the SDF-1/CXCR4 signaling pathway, facilitating tissue repair and regeneration .

  • Therapeutic delivery advances: Various delivery methods have been successfully employed in rat models, including gene transfer via electroporation for localized SDF-1β overexpression and direct local injection of recombinant protein. These approaches demonstrate the feasibility of SDF-1β-based therapeutic strategies .

  • Diverse cellular targets: SDF-1β affects multiple cell types beyond stem cells, including myofibroblasts, epithelial cells, and potentially inflammatory cells, highlighting its pleiotropic effects in complex biological systems .

  • Molecular signaling complexity: The effects of SDF-1β involve multiple downstream pathways and mediators, including TNF-α for myofibroblast apoptosis, suggesting intricate signaling networks that coordinate its diverse biological activities .

  • Translational potential: The positive outcomes observed in rat models of pulmonary fibrosis and bone regeneration provide a strong foundation for developing SDF-1β-based therapies for human diseases characterized by fibrosis or impaired tissue regeneration .

These key findings collectively highlight SDF-1β as a multifunctional signaling molecule with significant therapeutic potential across various disease contexts. The mechanistic insights gained from rat models provide critical guidance for both further preclinical research and potential clinical translation of SDF-1β-based interventions.

Product Science Overview

Structure and Isoforms

CXCL12 is part of the CXC chemokine family, characterized by the presence of four conserved cysteines that form two disulfide bonds. The initial pair of cysteines in CXC chemokines are separated by one intervening amino acid . The CXCL12 gene produces seven isoforms, with SDF-1α (CXCL12a) and SDF-1β (CXCL12b) being the most studied .

Expression and Function

CXCL12 is ubiquitously expressed in many tissues and cell types, including the brain, thymus, heart, lung, liver, kidney, spleen, platelets, and bone marrow . It is strongly chemotactic for lymphocytes and plays a significant role in the migration of hematopoietic cells from the fetal liver to the bone marrow during embryogenesis .

Signaling Pathways

CXCL12 signals through the G protein-coupled receptor CXCR4 to recruit activated leukocytes . This signaling pathway is involved in various cellular functions, including immune surveillance, inflammation response, tissue homeostasis, and tumor growth and metastasis .

Recombinant Rat CXCL12

Recombinant rat CXCL12/SDF-1β is a non-glycosylated protein composed of 72 amino acids with a molecular weight of 8.5 kDa . It is produced in response to inflammatory cytokines such as TNF, IL-1, or LPS . The recombinant form is used extensively in research to study its biological activities and potential therapeutic applications.

Biological Activity

The biological activity of CXCL12 includes its ability to chemoattract human peripheral T cells stimulated with IL-2 and PHA . This chemotactic property is crucial for its role in immune response and inflammation.

Clinical Relevance

CXCL12 and its receptor CXCR4 are involved in the regulation of migration, survival, and development of multiple cell types, including human hematopoietic stem cells . Additionally, CXCL12 signaling has been observed in several cancers, making it a target for cancer research and potential therapeutic interventions .

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.