SPARC Mouse

Secreted Protein Acidic & Rich in Cysteine Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Research Applications of SPARC Mouse Models

Key experimental uses include:

Ocular Physiology

  • SPARC knockout (SPARC -/-) mice exhibit 24% lower intraocular pressure (12.7 ± 0.6 mmHg vs 16.7 ± 0.9 mmHg in WT) due to enhanced aqueous humor outflow .

  • Associated with ECM remodeling in trabecular meshwork (40% reduction in fibronectin deposition) .

Metabolic Regulation

  • SPARC -/- mice develop 32% larger epididymal fat pads versus WT on standard diet :

ParameterWT MiceSPARC -/-P-value
Adipocyte diameter (μm)161 ± 33252 ± 610.03
Adipocytes/g tissue (x10³)49.8 ± 1.130.2 ± 0.9<0.01
Serum leptin (ng/mL)3.2 ± 0.46.8 ± 0.7<0.001

Cancer Biology

  • Lewis lung carcinoma tumors grow 4.8x faster in SPARC -/- hosts (1,930 mm³ vs 405 mm³ in WT) .

  • SPARC deficiency reduces tumor-infiltrating macrophages by 65% and VEGF receptor expression by 40% .

Hepatic Function

  • SPARC -/- mice on high-fat diets show:

    • 58% lower AST levels (124 U/L vs 295 U/L in WT)

    • 75% reduction in F4/80+ Kupffer cells

Mechanistic Insights from SPARC Studies

3.1 ECM Modulation
SPARC-null mice exhibit:

  • 50% reduction in dermal collagen I content

  • Disrupted lens capsule basement membrane integrity

  • Altered trabecular meshwork fibronectin/elastin ratios (3:1 vs 1.8:1 in WT)

Immune Interactions

  • SPARC+ myeloid-derived suppressor cells (MDSC) show 2.3x higher T-cell suppression versus SPARC-/- MDSC .

  • Tumors in SPARC -/- hosts exhibit 40% fewer PD-1+ exhausted CD8+ T-cells .

Therapeutic Implications

Disease AreaSPARC Modulation EffectReference
ObesityAdipocyte hyperplasia via ECM-collagen crosstalk
GlaucomaIOP reduction through MMP-mediated ECM remodeling
Hepatocellular carcinomaERK1/2-MMP2/9 pathway activation
NeuroinflammationAMPA receptor trafficking regulation

Technical Considerations

  • Recombinant SPARC bioactivity: ED₅₀ = 0.6-2.4 μg/mL in endothelial spreading assays

  • Antibody validation required due to SPARC's multiple cleavage forms (e.g., KGHK angiogenic peptide)

  • Strain-specific effects noted in C57Bl6/129SVJ vs BALB/c backgrounds

Product Specs

Introduction

Secreted Protein Acidic and Rich in Cysteine (SPARC), also known as osteonectin, is encoded by the SPARC gene in humans. This glycoprotein plays a crucial role in bone mineralization by binding to calcium. SPARC is synthesized by various cells, including fibroblasts, capillary endothelial cells, platelets, and macrophages, primarily in tissues undergoing morphogenesis and remodeling. Besides calcium, SPARC also exhibits binding affinity for collagen.

Description

Recombinant Mouse SPARC protein, expressed in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain comprising 291 amino acids (18-302a.a) with a molecular weight of 33.3kDa. This protein includes a 6 amino acid His-tag fused at the C-terminus and is purified using proprietary chromatographic techniques.

Physical Appearance
A sterile, filtered solution that is colorless.
Formulation

The SPARC solution is provided at a concentration of 0.25mg/ml and is formulated in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.

Stability

For short-term storage (2-4 weeks), the SPARC solution should be stored at 4°C. For extended storage, it is recommended to store the solution frozen at -20°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advisable. It is important to avoid repeated freeze-thaw cycles to maintain protein integrity.

Purity

The purity of SPARC Mouse Recombinant is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.

Synonyms

Osteonectin, ON, Basement-membrane protein 40, BM-40, SPARC, Secreted Protein acidic and Rich in Cysteine

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

APQQTEVAEE IVEEETVVEE TGVPVGANPV QVEMGEFEDG AEETVEEVVA DNPCQNHHCK HGKVCELDES NTPMCVCQDP TSCPAPIGEF EKVCSNDNKT FDSSCHFFAT KCTLEGTKKG HKLHLDYIGP CKYIAPCLDS ELTEFPLRMR DWLKNVLVTL YERDEGNNLL TEKQKLRVKK IHENEKRLEA GDHPVELLAR DFEKNYNMYI FPVHWQFGQL DQHPIDGYLS HTELAPLRAP LIPMEHCTTR FFETCDLDND KYIALEEWAG CFGIKEQDIN KDLVIHHHHH H

Q&A

What is SPARC and why are SPARC mouse models important for research?

SPARC (also known as osteonectin or BM-40) is a glycoprotein component of the extracellular matrix involved in diverse cellular processes. SPARC is produced by fibroblasts, capillary endothelial cells, platelets, and macrophages, particularly in areas undergoing tissue morphogenesis and remodeling . SPARC mouse models, especially SPARC-null (SPARC-/-) mice, provide valuable tools for investigating SPARC's role in normal development and pathological conditions. These models have revealed SPARC's involvement in collagen fiber assembly, cellular adhesion, migration, and various disease processes including cancer progression and intervertebral disc degeneration . Unlike conventional in vitro studies, these mouse models allow researchers to observe the systemic effects of SPARC deficiency in a living organism, providing insights that would be impossible to obtain through cell culture alone.

What are the primary phenotypic characteristics of SPARC-null mice?

SPARC-null mice exhibit several subtle but significant phenotypes that stem primarily from defective extracellular matrix assembly. While these mice are generally healthy and viable, they display distinct characteristics compared to wild-type counterparts . Most notably, SPARC-null mice show altered collagen fiber organization and assembly, as SPARC plays a critical role in proper collagen deposition and maturation. These mice are not inherently predisposed to spontaneous tumor formation . Age-dependent phenotypes include accelerated intervertebral disc degeneration, which makes these mice valuable models for studying low back pain pathology . Other reported phenotypic characteristics include alterations in wound healing, lens opacity, bone mineralization, and adipose tissue development. These phenotypes highlight SPARC's multifunctional role across various tissues and physiological processes.

How does SPARC expression vary across different tissues in mice?

SPARC expression demonstrates significant tissue specificity and developmental regulation in mice. During embryonic development, SPARC is prominently expressed in developing cartilage, as evidenced by immunohistochemical staining of mouse embryos (E15) . In adult mice, SPARC is predominantly expressed in tissues undergoing active remodeling or in cells associated with extracellular matrix production. Cell-specific expression has been documented in fibroblasts (such as Balb/C-3T3 cell line), myoblasts (C2C12 cell line), and placental tissue . Expression can also be detected in bone, skin, gastrointestinal tract, and central nervous system tissues. Notably, SPARC expression can be dynamically regulated in response to injury, inflammation, or disease states. This tissue-specific expression pattern reflects SPARC's context-dependent functions in different physiological and pathological processes.

What are the optimal methods for detecting SPARC expression in mouse tissues and cell lines?

Multiple complementary techniques provide robust detection of SPARC expression in mouse samples. Western blot analysis remains a gold standard for quantitative assessment, with optimal results obtained using specific antibodies such as Goat Anti-Mouse SPARC Antigen Affinity-purified Polyclonal Antibody at concentrations of 0.2 μg/mL . For subcellular localization studies, immunohistochemistry (IHC) and immunocytochemistry (ICC) deliver excellent results when using optimized fixation protocols. For frozen tissue sections, optimal conditions include overnight antibody incubation at 4°C followed by detection using HRP-DAB staining systems . For intracellular detection in cell cultures, flow cytometry requires proper fixation and permeabilization protocols, typically using specialized buffers to maintain cellular integrity while allowing antibody penetration . Researchers should validate detection methods using appropriate positive controls (such as C2C12 myoblasts or mouse placenta tissue) and negative controls (typically IgG isotype controls) to ensure specificity and minimize background signal.

How can researchers effectively generate and validate SPARC-knockout mouse models?

Generating reliable SPARC-knockout mouse models requires careful genetic engineering and thorough validation. While traditional knockout approaches using homologous recombination in embryonic stem cells have been historically employed (as in the original SPARC-/- mice generated by Norose et al. and described in multiple studies ), modern CRISPR/Cas9 techniques offer more efficient alternatives. Regardless of the methodology, comprehensive validation is essential and should include:

  • Genotyping PCR to confirm genetic modifications at the DNA level

  • Western blot analysis of tissue lysates to verify complete absence of SPARC protein (as demonstrated in the comparison of SPARC+/- and SPARC-/- TRAMP prostate and MMTV-PyMT mammary tumors )

  • Immunohistochemical staining of multiple tissues to confirm absence of protein expression

  • Phenotypic characterization to verify consistency with previously described SPARC-null phenotypes

  • Backcrossing to appropriate genetic backgrounds when introducing the knockout into specific disease models (as done with TRAMP and MMTV-PyMT models )

Researchers should maintain careful colony management with regular genotyping to prevent genetic drift and ensure experimental reproducibility.

What experimental controls are essential when studying SPARC in mouse disease models?

Rigorous experimental design with appropriate controls is critical for meaningful SPARC research. The minimum essential controls include:

  • Genetic controls: Include multiple genotypes (SPARC+/+, SPARC+/-, and SPARC-/-) to assess gene dosage effects, as demonstrated in studies comparing SPARC+/- and SPARC-/- mice in both TRAMP positive and negative backgrounds

  • Age-matched controls: SPARC-related phenotypes often show age-dependency, making age-matching crucial, especially in degenerative disease models like intervertebral disc degeneration

  • Sex-balanced groups: Include both male and female mice to account for potential sex-specific differences in SPARC function

  • Background strain controls: Maintain consistent genetic backgrounds or use appropriate congenic controls, particularly important when crossing SPARC-null mice with disease models like TRAMP (C57BL/6 background) or MMTV-PyMT (FVB background)

  • Technical controls for protein detection: Include positive controls (tissues known to express SPARC) and negative controls (isotype antibodies) for immunodetection methods

  • Disease progression controls: Include time-course analyses to differentiate between developmental defects and progressive pathological changes

This comprehensive control strategy ensures that observed phenotypes can be reliably attributed to SPARC alterations rather than experimental variables.

How does SPARC deficiency affect cancer progression and metastasis in mouse models?

The relationship between SPARC deficiency and cancer progression appears to be context-dependent and not universally pro- or anti-tumorigenic. Studies using SPARC-null mice crossed with transgenic cancer models have yielded important insights. In the TRAMP (transgenic adenocarcinoma of the mouse prostate) model, loss of SPARC surprisingly had no significant effect on tumor initiation, progression, or metastasis rates. Similarly, in the MMTV-PyMT (murine mammary tumor virus-polyoma middle T) breast cancer model, SPARC deficiency did not substantially alter tumor development or progression .

These findings contrast with some previous assumptions that altered SPARC expression would significantly impact cancer progression. In both models, neither the incidence of palpable carcinomas (35% versus 28% in TRAMP models) nor macroscopic lymph node metastases (25% versus 21%) differed significantly between SPARC+/- and SPARC-/- mice . Additionally, tumor angiogenesis and collagen deposition were largely unaffected by SPARC status. These results suggest that while SPARC expression levels may serve as useful biomarkers for aggressive tumors, SPARC deficiency alone may not be sufficient to promote or inhibit cancer progression in certain spontaneous mouse tumor models.

What role does SPARC play in intervertebral disc degeneration and low back pain models?

SPARC deficiency significantly impacts intervertebral disc (IVD) integrity and is associated with accelerated disc degeneration and pain development. SPARC-null mice serve as valuable models for studying chronic low back pain associated with IVD pathology. These mice exhibit age-dependent behavioral signs of chronic axial low back pain and radiating leg pain that correlate with progressive disc degeneration .

The pathophysiological mechanism involves impaired extracellular matrix maintenance in IVD tissues, leading to structural deterioration. This degeneration triggers sensory nervous system plasticity, including increased innervation of spinal tissues. The SPARC-null mouse model has proven particularly valuable for investigating the neurobiological basis of discogenic pain, as it recapitulates key aspects of human degenerative disc disease . Immunohistochemical and behavioral studies using this model have revealed important insights into the relationship between disc structural changes, neuronal plasticity, and pain behaviors. This model offers advantages over induced injury models by providing a progressive, age-related degeneration pattern that more closely mimics the human clinical condition.

How is SPARC involved in fibrotic disorders and tissue remodeling in mouse models?

SPARC plays a significant role in fibrotic processes and tissue remodeling through its effects on extracellular matrix organization and cellular behavior. Recent research has implicated SPARC in keloid formation, with significantly elevated SPARC expression observed in keloid tissues compared to normal skin tissues . This suggests SPARC's involvement in the excessive collagen deposition and abnormal wound healing characteristic of keloids.

In mouse models, SPARC influences fibrotic responses through multiple mechanisms:

  • Regulation of collagen fiber assembly and organization

  • Modulation of growth factor signaling, particularly TGF-β pathways that drive fibroblast activation

  • Influence on matrix metalloproteinase (MMP) expression and activity

  • Promotion of cellular migration during tissue remodeling

  • Mediation of intermediate adhesion states that facilitate cellular reorganization

SPARC's functional role extends to multiple fibrotic conditions beyond keloids, including liver fibrosis, pulmonary fibrosis, and cardiac fibrosis. The activation of specific signaling pathways, such as p38γ signaling, appears important for SPARC's pro-fibrotic effects, as demonstrated in recent studies showing that SPARC can activate this pathway to stabilize metabolic enzymes like PFKFB3 and promote glycolysis in fibroblasts .

What are the key signaling pathways mediated by SPARC in mouse development and disease?

SPARC engages multiple signaling cascades that influence cellular behavior in both developmental and pathological contexts. The protein functions through both direct cellular receptor interactions and indirect effects on extracellular matrix organization and growth factor availability. Key signaling pathways include:

  • Integrin-mediated signaling: SPARC modulates integrin-mediated adhesions, thereby affecting focal adhesion dynamics and downstream signaling molecules like FAK (Focal Adhesion Kinase)

  • TGF-β pathway: SPARC can induce and/or activate TGF-β signaling, contributing to its effects on cellular differentiation, migration, and matrix production

  • MAPK pathways: Recent evidence indicates SPARC activates specific MAPK family members, particularly p38γ, which can regulate metabolic enzymes like PFKFB3 and influence cellular metabolism

  • MMP regulation: SPARC influences the expression and activation of matrix metalloproteinases, affecting matrix remodeling and cellular invasion

  • Cytokine/growth factor availability: By binding to various matrix substrates and growth factors, SPARC can modulate their availability to cells

These signaling mechanisms explain SPARC's diverse biological effects, including its ability to reorganize stress fibers, promote intermediate adhesion states favorable for migration, and influence collagen fiber assembly.

How do SPARC interactions with the extracellular matrix influence cell behavior in mouse models?

SPARC functions as a critical matricellular protein that mediates bidirectional communication between cells and their extracellular environment. In mouse models, these interactions manifest through several mechanisms:

  • Collagen binding and organization: SPARC directly binds collagen and is essential for proper collagen fiber assembly, as evidenced by altered collagen architecture in SPARC-null mice

  • Focal adhesion modulation: SPARC promotes disassembly of cellular focal adhesions and reorganization of actin stress fibers, creating an "intermediate" adhesion state that facilitates cell migration

  • ECM-growth factor crosstalk: SPARC binds various matrix components like collagen and laminin, potentially affecting their interaction with growth factors and cytokines

  • Cell-surface receptor interactions: Though specific SPARC receptors remain somewhat controversial, SPARC interacts with cellular receptors to influence intracellular signaling

  • Tissue-specific matrix composition: SPARC deficiency affects ECM composition differently across tissues, explaining why phenotypes in SPARC-null mice may be more prominent in certain tissues

In neuronal contexts, SPARC influences AMPA receptor availability and function, demonstrating its diverse impact on cellular behavior beyond traditional matrix-related functions. Studies show that SPARC-containing AMPA receptors promote neuronal health following CNS injury , highlighting the diverse roles of SPARC in different tissue environments.

What are the molecular mechanisms underlying SPARC's role in inflammation and immune responses?

SPARC exerts complex effects on inflammatory processes and immune cell function through both direct and indirect mechanisms:

  • Macrophage production and function: SPARC is produced by macrophages, particularly in areas of tissue remodeling, suggesting autocrine and paracrine functions in these immune cells

  • Inflammatory pathway modulation: Recent evidence indicates SPARC can activate inflammatory signaling cascades, including specific MAPK pathways like p38γ

  • ECM-mediated immune regulation: SPARC's effects on ECM organization indirectly influence immune cell migration, adhesion, and activation

  • Tissue-specific inflammatory effects: In acute pancreatitis models, SPARC ablation modifies disease activity, suggesting context-specific roles in inflammatory conditions

  • Crosstalk with cytokine networks: SPARC interacts with various cytokines and may regulate their availability and signaling potential

  • Myeloid cell effects: CSF1-ETS2-induced microRNA in myeloid cells promotes metastatic tumor growth, with SPARC potentially involved in this pathway

These mechanisms highlight SPARC's multifaceted role in inflammatory processes, explaining its involvement in conditions ranging from cancer to fibrotic disorders. The precise effects appear highly context-dependent, varying with tissue type, disease state, and the specific immune cell populations involved.

What are the optimal experimental approaches for studying SPARC function in specific mouse tissues?

Tissue-specific SPARC function requires tailored experimental approaches that account for its context-dependent activities:

  • For nervous system studies: Combine behavioral assessments with immunohistochemical analyses to correlate SPARC expression with functional outcomes. This approach has proven valuable in studies of SPARC's role in convulsive severity following pentylenetetrazol injection in schizencephaly and microgyria models . Similarly, examining developmental expression patterns of SPARC in the Fragile X mouse model has yielded insights into neurodevelopmental disorders .

  • For metabolic tissue analysis: Integrate tissue-specific knock-out/overexpression systems with comprehensive metabolic phenotyping. This approach revealed that SPARC knockout led to an accelerated aging phenotype that could be improved by exercise, while SPARC overexpression mimicked exercise effects .

  • For cancer studies: Cross SPARC-null mice with established tumor models like TRAMP (prostate cancer) or MMTV-PyMT (breast cancer) to evaluate effects on tumor initiation, progression, and metastasis . Complementary approaches should include angiogenesis assessment, collagen deposition analysis, and immune infiltration characterization.

  • For inflammatory conditions: Use conditional expression systems to modulate SPARC levels at specific disease stages. This approach revealed SPARC's role in colorectal cancer, where USP22 suppresses SPARC expression in acute colitis and inflammation-associated colorectal cancer .

Each experimental approach should incorporate appropriate controls, temporal analyses, and multiple methodological approaches to provide comprehensive insights into SPARC's tissue-specific functions.

How can SPARC mouse models advance translational research for human diseases?

SPARC mouse models offer significant translational value for understanding and treating human diseases:

  • Degenerative disc disease: The SPARC-null mouse serves as an excellent model of age-dependent intervertebral disc degeneration and associated pain behaviors that closely mimic human pathology . This model enables testing of novel analgesics, regenerative therapies, and biological interventions before clinical trials.

  • Cancer therapy development: Although SPARC deficiency alone may not significantly alter tumor progression in some models , understanding context-specific SPARC functions can help identify patient subpopulations most likely to benefit from SPARC-targeted therapies or predict treatment responses.

  • Fibrotic disorders: The elevated SPARC expression observed in keloid tissues points to potential therapeutic approaches targeting SPARC or its downstream effectors . Mouse models allow testing of anti-fibrotic interventions before human application.

  • Aging and metabolism: The accelerated aging phenotype in SPARC-null mice that improves with exercise suggests SPARC might represent a molecular link between exercise and anti-aging effects , offering potential therapeutic targets for age-related conditions.

  • Neurological conditions: SPARC's role in promoting neuronal health following CNS injury through GluA1-containing AMPA receptors suggests potential neuroprotective strategies for traumatic brain injury, stroke, and neurodegenerative diseases.

These translational applications highlight the value of SPARC mouse models for bridging basic science discoveries with potential clinical innovations.

What emerging technologies can enhance SPARC research in mouse models?

Several cutting-edge technologies are transforming SPARC research in mouse models:

  • Single-cell RNA sequencing: This technology allows researchers to examine cell-specific SPARC expression patterns and responses, providing unprecedented resolution of SPARC's role in heterogeneous tissues. This approach could resolve contradictory findings by identifying cell-type-specific functions that might be masked in whole-tissue analyses.

  • CRISPR-based in vivo gene editing: Beyond traditional knockout approaches, CRISPR systems enable tissue-specific and temporally controlled SPARC modulation in adult mice. This overcomes limitations of conventional knockout models where developmental adaptations might mask acute SPARC functions.

  • Advanced imaging techniques: Techniques like intravital microscopy allow real-time visualization of SPARC-dependent processes in living tissues. Combined with fluorescent reporter systems, researchers can track dynamic changes in SPARC expression and localization during disease progression.

  • Spatial transcriptomics and proteomics: These approaches can map SPARC expression and its effects on other genes and proteins with spatial context preserved, providing insights into localized SPARC functions within complex tissues.

  • Organoid models derived from SPARC-modified mice: These "mini-organs" enable controlled experiments in systems that better recapitulate the complexity of in vivo tissues while offering experimental accessibility.

  • Computational modeling of SPARC-ECM interactions: Advanced simulations can predict how SPARC modifications alter extracellular matrix properties and subsequent cellular behaviors, guiding experimental design.

These technological advances promise to resolve existing questions about SPARC function and open new avenues for understanding its complex roles in development and disease.

Product Science Overview

Structure and Composition

SPARC is a 40 kDa acidic and cysteine-rich glycoprotein consisting of a single polypeptide chain. It can be divided into four domains:

  1. Ca²⁺ Binding Domain: Located near the glutamic acid-rich region at the amino terminus.
  2. Cysteine-Rich Domain: Contains multiple cysteine residues.
  3. Hydrophilic Region: A region that interacts with water molecules.
  4. EF Hand Motif: Located at the carboxy terminus, involved in calcium binding .
Functions

SPARC is involved in several biological processes, including:

  • Bone Mineralization: It binds to calcium and collagen, promoting the formation of mineral crystals in bones .
  • ECM Remodeling: Acts as a matricellular chaperone, mediating the disassembly and degradation of ECM networks .
  • Cell-Matrix Interactions: Modulates interactions between cells and the ECM, influencing cell adhesion, migration, and proliferation .
  • Tumor Biology: Overexpression of SPARC has been linked to various cancers, including breast, prostate, colon, and pancreatic cancers. It promotes angiogenesis, proliferation, and migration of tumor cells .
Mouse Recombinant SPARC

The mouse recombinant version of SPARC is produced using recombinant DNA technology, where the SPARC gene from mice is inserted into a suitable expression system, such as bacteria or yeast. This allows for the production of large quantities of SPARC protein for research and therapeutic purposes.

Research and Applications

SPARC has been extensively studied for its role in tissue remodeling and repair. It has potential therapeutic applications in treating diseases related to ECM dysfunction, such as fibrosis and cancer . Additionally, SPARC’s role in bone mineralization makes it a target for osteoporosis research .

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.