SPOCK3 Mouse

Sparc/Osteonectin 3 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Nomenclature

SPOCK3, symbolized as spock3 in mice, is a proteoglycan encoded by the SPARC (osteonectin), cwcv and kazal like domains proteoglycan 3 gene. It belongs to the BM-40/SPARC/osteonectin family, which regulates extracellular matrix (ECM) dynamics and cellular interactions . In mice, SPOCK3 is also referred to as Testican-3 due to its structural homology with other Testican family members .

Expression Patterns and Localization

SPOCK3 expression in mice is developmentally regulated and tissue-specific:

Developmental Stages

StageKey Tissues/Organs
EmbryonicVascular system, liver, inner ear, central nervous system (CNS)
AdultConfined to the brain, particularly thalamic nuclei, olfactory bulb, cortex, hippocampus, and striatum

Neuroanatomical Distribution

In adult mice, SPOCK3 is abundant in regions critical for sensory processing and behavior:

  • Thalamus: Regulates sensory input and relay

  • Hippocampus: Involved in memory formation

  • Striatum: Modulates motor control and reward pathways

Neurodevelopmental Role

Studies using Spock3-mutant mice reveal severe structural and behavioral deficits:

PhenotypeObservation
Corpus callosum morphologyNarrowing and myelin sheath defects, particularly in the central region
Cortical axon organizationDisrupted bundling of input/output fibers; misrouting into dorsal striatum
Behavioral outcomesReduced anxiety-like behavior and sociability

These findings highlight SPOCK3’s role in maintaining axonal integrity and synaptic connectivity during brain development .

Tumor Biology and Protease Inhibition

SPOCK3 modulates tumor progression through:

  • MMP-2 Inhibition: The N-terminal region inhibits MT1-MMP/MT3-MMP-mediated activation of pro-MMP-2, suppressing ECM degradation

  • Prognostic Biomarker: High expression correlates with improved disease-free survival in prostate cancer, while downregulation in gliomas reduces cell migration

Recombinant SPOCK3 Proteins and Applications

Commercially available recombinant SPOCK3 proteins facilitate functional studies:

SourceAmino Acid RangePurityTagApplications
Baculovirus (Boster)22–436>95%His-tagSDS-PAGE, HPLC
HEK 293 (Abcam)23–436>95%His-tagSDS-PAGE, inhibition assays
E. coli (R&D Systems)26–436Carrier-freeHis-tagCathepsin L inhibition assays

These proteins are used to study protease interactions (e.g., cathepsin L, MMP-14) and ECM remodeling .

Product Specs

Introduction
Testican-3, also known as Spock3, is a heparan sulfate proteoglycan found in the nervous system. Part of the BM-40/SPARC/osteonectin family, its exact role in brain development remains unclear. Spock3 is known to inhibit the processing of pro-matrix metalloproteinase 2 (MMP-2) by MT1-MMP and MT3-MMP. Its presence is primarily observed during the developmental stage of the brain.
Description
This product consists of recombinant SPOCK3 protein derived from mice. Produced in Sf9 Baculovirus cells, it exists as a single, glycosylated polypeptide chain with a molecular weight of 47.9kDa. The protein sequence encompasses amino acids 22-436 and includes an 8 amino acid His tag at the C-terminus. Purification is achieved using proprietary chromatographic techniques. On SDS-PAGE under reducing conditions, the protein migrates between 40-57kDa.
Physical Appearance
A clear and colorless solution, sterilized by filtration.
Formulation
The SPOCK3 protein solution has a concentration of 0.25mg/ml and is supplied in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. To maintain product integrity, avoid repeated freeze-thaw cycles.
Purity
Purity of the SPOCK3 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Testican-3, SPARC/osteonectin, CWCV, Kazal-like domains proteoglycan 3, Spock3.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
AAAAVAVAGG RSDGGNFLDE KQWLTTISQY DKEVGQWNKF RDEVEDDYFR TWNPGKPFDQ ALDPAKDPCL KTKCSRHKVC ITQDAQTALC ISHRRLTHSM KEVGGSHKQW RGLPSSTCKP CPIAYASPVC GSDGHSYSSQ CKLEYQACVL GKQISIKCEG RCPCPSDKSM NIGRNVKRAC
SDLEFREVAN RLRDWFKALH ESGSQNKKTK ALLRPERSRF DTSILPICKD SLGWMFNRLD TNYDLLLDQS ELGSIYLDKN EQCTKAFFNS CDTYKDSLIS NNEWCYCFQR QQDPPCHTEL SNIQKRQGIK KLLGQYIPLC DEDGYYKPTQ CHGSVGQCWC VDRYGNEVVG SRINGVADCA
IDFEISGDFA SGDFREWTDD EGEEDDIMND KDDIEDDDED EGDDDDDGDV HDGYILEHHH HHH

Q&A

What is the molecular structure of mouse SPOCK3 protein?

Mouse SPOCK3 is a glycosylated polypeptide containing 423 amino acids (residues 22-436) with a molecular mass of approximately 47.9kDa, though it typically migrates at 40-57kDa on SDS-PAGE under reducing conditions due to post-translational modifications . The protein contains several functional domains including calcium-binding sites (EF-hand motifs), Kazal domains, and thyroglobulin type-1 domains . SPOCK3 undergoes significant post-translational modifications, containing both chondroitin sulfate and heparan sulfate O-linked oligosaccharides that are critical for its functionality in the extracellular matrix .

What are the primary biological functions of SPOCK3 in mice?

SPOCK3 participates in diverse steps of neurogenesis and plays a crucial role in the formation and maintenance of major neuronal structures in the brain . Functionally, it inhibits the processing of pro-matrix metalloproteinase 2 (MMP-2) by MT1-MMP and MT3-MMP, potentially interfering with tumor invasion mechanisms . Research with SPOCK3-mutant mice has demonstrated that the protein is essential for proper development of cortical layers and corpus callosum formation . Additionally, SPOCK3 appears to be involved in regulating anxiety-like behavior and sociability, as mutant mice exhibit decreased anxiety-like behavior and lowered social interaction .

How does SPOCK3 differ from other members of the SPOCK family in expression and function?

While all SPOCK family members (SPOCK1, SPOCK2, and SPOCK3) are expressed during brain development, their expression patterns diverge significantly in adult mice. SPOCK1 and SPOCK2 maintain intense expression throughout the entire adult brain, whereas SPOCK3 expression becomes highly restricted, remaining detectable primarily in thalamic nuclei . This suggests that SPOCK3's function is mostly confined to the developmental stage of the brain, unlike its family members that continue to play roles in the adult nervous system . Functionally, SPOCK1 is known to inhibit cell attachment and neurite outgrowth in cultured neuronal cells, and SPOCK3 was initially thought to function similarly, though subsequent research has revealed its more specific role in axonal tract formation and maintenance .

How can researchers effectively measure SPOCK3 expression in mouse tissues?

For measuring SPOCK3 expression in mouse tissues, researchers can employ multiple complementary approaches:

  • mRNA detection: RT-PCR or in situ hybridization can identify Spock3 transcripts in tissue samples. Multiple mRNA variants (spock3-201 through spock3-205) have been identified, ranging from 971 to 2,459 nucleotides in length .

  • Protein detection: Commercial antibodies specifically targeting mouse SPOCK3 are available for immunohistochemistry, western blotting, or ELISA . When performing western blot analysis, researchers should note that SPOCK3 typically migrates at 40-57kDa on SDS-PAGE under reducing conditions despite its calculated molecular mass of 47.9kDa .

  • Recombinant protein standards: Purified recombinant mouse SPOCK3 protein (>95% purity) can be used as positive controls in expression studies .

For optimal results, tissue preparation should account for SPOCK3's extracellular matrix localization, and appropriate extraction methods should be employed to isolate proteoglycans effectively.

What factors regulate SPOCK3 expression in mouse models?

The regulation of SPOCK3 expression appears complex and highly developmentally controlled. While specific transcriptional regulators have not been fully characterized in the provided search results, the strict developmental regulation of SPOCK3—with high expression during brain development followed by restricted expression in adulthood—suggests sophisticated transcriptional control mechanisms . The SPOCK3 gene contains multiple transcript variants (five documented variants in zebrafish: spock3-201 through spock3-205), indicating potential alternative splicing regulation . Researchers investigating SPOCK3 regulation should consider examining developmental signaling pathways associated with neurogenesis and axonal guidance, as these likely influence SPOCK3 expression patterns. Additionally, studies utilizing SPOCK3-mutant mice with deletions extending from the upstream regulatory region to exon 4 have demonstrated the significance of these genomic elements in controlling proper SPOCK3 expression .

What neuroanatomical abnormalities are observed in SPOCK3-deficient mice?

SPOCK3-deficient mice exhibit several distinct neuroanatomical abnormalities:

  • Cortical abnormalities: Cells in all cortical layers appear swollen compared to wild-type counterparts .

  • Corpus callosum defects: The corpus callosum shows significant narrowing around the central region along the rostral-caudal axis, with many small spaces observed throughout the structure lacking myelin sheaths .

  • Fiber tract disorganization: Both cortical input and output fibers fail to form thick bundled fibers as observed in wild-type mice .

  • Corticospinal tract abnormalities: A subpopulation of corticospinal axonal fibers penetrates into the dorsal striatum with moderately altered orientations, indicating errors in axonal pathfinding or maintenance .

These structural abnormalities are consistent with behavioral changes observed in these mice, including decreased anxiety-like behavior and reduced sociability . This suggests SPOCK3 plays a critical role in the proper formation or maintenance of major neuronal structures that control complex behaviors.

How does SPOCK3 influence axonal guidance and myelination?

SPOCK3 appears to play crucial roles in axonal guidance and possibly myelination through several potential mechanisms:

SPOCK3 may influence axonal guidance through:

  • Regulation of extracellular matrix composition, as it functions as a proteoglycan component of the ECM

  • Modulation of metalloproteinase activity, specifically inhibiting pro-MMP2 processing by MT1-MMP and MT3-MMP, which may maintain ECM integrity during axonal navigation

  • Potential interaction with guidance cues, as suggested by the altered trajectory of corticospinal axonal fibers in SPOCK3-deficient mice

Regarding myelination, SPOCK3-deficient mice exhibit spaces throughout the corpus callosum lacking myelin sheaths . This suggests SPOCK3 may influence either:

  • Oligodendrocyte development or function

  • The stabilization of axon-myelin interactions

  • The creation of a permissive extracellular environment for proper myelination

Research methodologies to further investigate these roles would include detailed immunohistochemical analysis of developing axonal tracts in SPOCK3-deficient versus wild-type mice, in vitro axonal guidance assays using neurons cultured on substrates containing or lacking SPOCK3, and electron microscopic examination of myelin structure in affected regions.

What behavioral phenotypes are associated with SPOCK3 mutations in mice?

SPOCK3-mutant mice exhibit specific behavioral phenotypes that correlate with the neuroanatomical abnormalities observed:

  • Decreased anxiety-like behavior: Mutant mice show reduced anxiety responses in behavioral testing paradigms .

  • Lowered sociability: SPOCK3-deficient mice demonstrate diminished social interaction behaviors compared to wild-type counterparts .

These behavioral alterations are consistent with the structural abnormalities observed in the corpus callosum and cortical axonal tracts, which are known to be involved in emotional regulation and social behavior processing . For researchers studying SPOCK3's role in behavior, standardized tests including elevated plus maze, open field tests, social preference assays, and cognitive assessment batteries would be recommended to fully characterize the behavioral phenotype. Additionally, correlating specific neuroanatomical defects with behavioral outcomes using techniques such as tract-tracing combined with behavioral analysis could provide valuable insights into the circuit-level mechanisms by which SPOCK3 influences behavior.

What are the optimal methods for generating SPOCK3 knockout or mutant mouse models?

Based on successful previous research, optimal methods for generating SPOCK3 knockout or mutant mouse models include:

  • Targeted deletion strategy: The most well-documented approach involves creating a deletion extending from the presumptive upstream regulatory region to exon 4 of the SPOCK3 locus . This strategy effectively eliminates SPOCK3 expression while allowing for the study of resulting phenotypes.

  • CRISPR/Cas9 genome editing: While not explicitly mentioned in the search results, this contemporary approach would allow for precise targeting of the SPOCK3 gene. Target sites should be designed within early exons (particularly exons 1-4) to ensure complete functional disruption.

  • Conditional knockout approach: For researchers interested in tissue-specific or temporally controlled SPOCK3 deletion, a Cre-loxP system targeting critical exons would be optimal. This would be particularly valuable given SPOCK3's developmental expression pattern and its restricted expression in adult thalamic nuclei .

When generating these models, researchers should include appropriate controls and validate the knockout by confirming absence of SPOCK3 protein using Western blot analysis and immunohistochemistry, particularly in neuronal tissues where SPOCK3 is normally expressed .

What are the recommended protocols for purifying recombinant mouse SPOCK3 protein?

For purifying recombinant mouse SPOCK3 protein, the following protocol synthesis based on established methods is recommended:

  • Expression systems: Two effective expression systems have been documented:

    • HEK 293 cells: Produce Full Length SPOCK3 protein (aa 23-436) with >95% purity

    • Sf9 Baculovirus cells: Generate glycosylated SPOCK3 (aa 22-436) with molecular mass of 47.9kDa

  • Affinity tags: Including an 8-amino acid His tag at the C-terminus facilitates purification .

  • Purification steps:

    • Initial capture: Nickel or cobalt affinity chromatography for His-tagged protein

    • Refinement: Proprietary chromatographic techniques as referenced in commercial preparations

  • Quality control:

    • SDS-PAGE analysis: Verify purity >90%, noting that SPOCK3 typically migrates at 40-57kDa despite its calculated mass

    • Endotoxin testing: Ensure levels <1 EU/μg

    • Functional validation: Verify calcium-binding activity and MMP-2 inhibition properties

  • Storage formulation: Optimal storage in phosphate-buffered saline (pH 7.4) with 10% glycerol .

This protocol synthesis provides a framework for researchers seeking to purify functional mouse SPOCK3 for experimental applications.

How can researchers effectively study SPOCK3 interactions with matrix metalloproteinases?

To effectively study SPOCK3 interactions with matrix metalloproteinases (MMPs), researchers should employ multiple complementary approaches:

  • In vitro inhibition assays:

    • Measure pro-MMP-2 processing by MT1-MMP and MT3-MMP in the presence vs. absence of purified SPOCK3

    • Use zymography to assess MMP activity with varying concentrations of SPOCK3

    • Employ fluorogenic substrates to quantify MMP catalytic activity when modulated by SPOCK3

  • Binding studies:

    • Surface plasmon resonance (SPR) to determine binding kinetics between SPOCK3 and MMPs

    • Co-immunoprecipitation experiments from neural tissue or transfected cells

    • FRET-based interaction assays for real-time monitoring of SPOCK3-MMP binding

  • Domain mapping:

    • Generate SPOCK3 constructs with specific domain deletions to identify regions critical for MMP interaction

    • Focus on thyroglobulin type-1 and Kazal domains which may mediate protease inhibition

  • Cell-based systems:

    • Transfect neuronal cell lines with SPOCK3 and assess changes in MMP activity and localization

    • Use SPOCK3-deficient primary neurons from knockout mice to examine altered MMP processing

  • In vivo validation:

    • Compare MMP activity and processing in tissues from wild-type versus SPOCK3-deficient mice

    • Employ in vivo imaging of MMP activity in SPOCK3 mutant models

This multi-faceted approach would provide comprehensive insights into the molecular mechanisms of SPOCK3's inhibition of MMP processing and its functional consequences in neural development.

What is the potential role of SPOCK3 in neurological disorders based on mouse model studies?

Based on the phenotypic analysis of SPOCK3-deficient mice, several potential roles in neurological disorders can be inferred:

  • Corpus callosum disorders: The narrowed corpus callosum with unmyelinated spaces observed in SPOCK3-mutant mice suggests SPOCK3 may be relevant to corpus callosum dysgenesis or hypoplasia in humans. Researchers should investigate SPOCK3 variants in patients with these conditions.

  • Social behavior disorders: Given the reduced sociability in SPOCK3-deficient mice , there may be implications for disorders characterized by social impairments, such as autism spectrum disorders. Research methodologies should include detailed behavioral phenotyping using social cognition tasks.

  • Anxiety disorders: The decreased anxiety-like behavior in mutant mice suggests SPOCK3 may modulate anxiety-related circuits. This could be relevant to both anxiety disorders and conditions with altered anxiety responses.

  • White matter pathologies: The myelin abnormalities in SPOCK3-deficient mice indicate potential relevance to demyelinating or dysmyelinating disorders. Further investigation using more detailed myelin imaging and biochemical analysis is warranted.

  • Neurodevelopmental disorders: The critical role of SPOCK3 in brain development suggests its dysfunction could contribute to broader neurodevelopmental disorders. Researchers should consider SPOCK3 sequencing in cohorts with these conditions.

Methods to further explore these connections include comparative transcriptomics between mouse models and human patient samples, detailed circuit analysis of affected pathways, and pharmacological rescue experiments targeting pathways downstream of SPOCK3.

How might SPOCK3 be involved in tumor invasion and metastasis based on its MMP inhibitory properties?

SPOCK3's documented ability to inhibit the processing of pro-matrix metalloproteinase 2 (MMP-2) by MT1-MMP and MT3-MMP suggests several potential mechanisms by which it might influence tumor invasion and metastasis:

  • Extracellular matrix remodeling: By inhibiting MMP activation, SPOCK3 may prevent the degradation of ECM components that typically create pathways for tumor cell invasion .

  • Tumor microenvironment regulation: SPOCK3 could modify the tumor microenvironment by preserving ECM integrity, potentially limiting the space for tumor expansion and migration.

  • Growth factor availability: MMPs often release matrix-bound growth factors; SPOCK3's inhibition of MMP activity might restrict the availability of these factors, limiting tumor growth signals.

  • Cancer cell motility: The matrix-preserving effects of SPOCK3 might create a less permissive environment for cancer cell motility, potentially reducing metastatic spread.

To investigate these mechanisms, researchers should consider:

  • Comparing SPOCK3 expression levels across various cancer types and correlating with invasiveness

  • Performing in vitro invasion assays with cancer cells in the presence/absence of SPOCK3

  • Developing orthotopic tumor models in SPOCK3-deficient mice to assess changes in tumor behavior

  • Evaluating SPOCK3 as a potential biomarker for tumor progression or therapeutic target

These approaches would help clarify SPOCK3's role in regulating tumor behavior through its interaction with the MMP system and potential applications in cancer research.

What is the comparative expression of SPOCK3 across different mouse strains commonly used in research?

Although the search results don't provide direct comparative data on SPOCK3 expression across different mouse strains, researchers interested in this question should consider the following methodological approach to generate this valuable information:

  • Strain selection: Include diverse commonly used laboratory mouse strains such as:

StrainCoat colorCommon research applications
C57BL/6JBlackGeneral purpose, background for most transgenics
BALB/cByJAlbinoCancer, cardiovascular, immunology
DBA/2JDilute brownCardiovascular, developmental biology
C3H/HeJAgoutiCancer, cardiovascular, hematology
129 strainsVariousStem cell research, knockout models
  • Multi-tissue expression analysis: Compare SPOCK3 expression across brain regions with particular attention to:

    • Developmental timepoints (embryonic, postnatal, adult)

    • Thalamic nuclei (where SPOCK3 expression persists in adults)

    • Areas relevant to observed phenotypes (corpus callosum, cortical layers)

  • Methodology recommendations:

    • Quantitative RT-PCR for precise measurement of mRNA levels

    • RNAscope for spatial resolution of expression patterns

    • Western blotting with strain-matched loading controls

    • Immunohistochemistry with quantitative image analysis

  • Data normalization: Account for baseline differences in brain size and cellularity between strains

This comparative analysis would provide important insights into strain-dependent variations that might influence SPOCK3-related phenotypes and guide appropriate strain selection for SPOCK3 research.

How can SPOCK3 mouse models be utilized to investigate axonal regeneration after injury?

SPOCK3 mouse models offer valuable tools for investigating axonal regeneration after injury, particularly given SPOCK3's roles in axonal tract formation and maintenance . A comprehensive research approach would include:

  • Injury models in SPOCK3-deficient mice:

    • Spinal cord injury paradigms to assess corticospinal tract regeneration

    • Optic nerve crush models to evaluate retinal ganglion cell axon regeneration

    • Peripheral nerve injury (sciatic nerve) to compare central vs. peripheral regeneration

  • Intervention approaches:

    • SPOCK3 supplementation: Delivery of recombinant SPOCK3 to injury sites to assess potential pro-regenerative effects

    • Domain-specific interventions: Administration of specific SPOCK3 functional domains to determine which regions mediate regenerative effects

    • Combinatorial approaches: SPOCK3 modulation alongside established pro-regenerative interventions

  • Assessment methodologies:

    • Anterograde and retrograde tracing of regenerating axons

    • Live imaging of labeled axons in transparent tissue preparations

    • Functional recovery assessment using behavioral and electrophysiological measures

    • Molecular analysis of regeneration-associated gene expression

  • Mechanistic investigations:

    • Evaluation of MMP activity in the injury site with and without SPOCK3

    • Analysis of growth cone dynamics on SPOCK3-containing substrates

    • Assessment of SPOCK3's influence on myelin-associated inhibitory factors

This research paradigm would leverage the unique features of SPOCK3 mouse models to potentially uncover novel approaches to enhancing axonal regeneration after injury.

What experimental approaches can determine SPOCK3's role in synaptic plasticity and function?

To investigate SPOCK3's potential role in synaptic plasticity and function, researchers should implement a multi-level experimental approach:

  • Electrophysiological analyses:

    • Perform patch-clamp recordings in brain slices from SPOCK3-deficient vs. wild-type mice

    • Assess long-term potentiation (LTP) and depression (LTD) in regions with developmental SPOCK3 expression

    • Evaluate synaptic transmission parameters including paired-pulse facilitation, miniature synaptic currents, and evoked responses

  • Structural analyses:

    • Utilize high-resolution microscopy (electron microscopy, super-resolution techniques) to examine synaptic ultrastructure

    • Quantify dendritic spine density, morphology, and turnover through longitudinal in vivo imaging

    • Assess synaptic protein composition and organization using array tomography or expansion microscopy

  • Molecular approaches:

    • Perform synaptic proteomics comparing wild-type and SPOCK3-deficient synaptosomal preparations

    • Investigate SPOCK3 localization relative to synaptic markers using high-resolution imaging

    • Examine activity-dependent regulation of SPOCK3 expression or localization

  • Behavioral correlates:

    • Assess learning and memory paradigms dependent on synaptic plasticity

    • Utilize experience-dependent plasticity paradigms (e.g., monocular deprivation)

    • Correlate electrophysiological findings with behavioral outcomes

  • Rescue experiments:

    • Attempt restoration of normal plasticity through viral delivery of SPOCK3 to deficient animals

    • Perform time-dependent interventions to identify critical developmental windows

This comprehensive approach would help elucidate whether SPOCK3's documented effects on brain structure have functional consequences for synaptic transmission and plasticity, potentially expanding understanding of its neurobiological roles.

What are the current technical challenges in studying SPOCK3 function and potential strategies to overcome them?

Researchers face several technical challenges when studying SPOCK3 function, along with potential strategies to address them:

  • Challenge: Distinguishing direct SPOCK3 effects from developmental consequences
    Solution: Employ conditional knockout systems (Cre-loxP) allowing for temporally controlled deletion of SPOCK3 at different developmental stages or in adulthood . This approach would help separate direct functional effects from developmental structural abnormalities.

  • Challenge: SPOCK3's complex post-translational modifications
    Solution: Utilize expression systems capable of proper glycosylation (e.g., mammalian cells rather than bacterial systems) . Consider domain-specific studies to isolate functions independent of glycosylation state.

  • Challenge: Limited expression in adult tissue complicating functional studies
    Solution: Focus studies on thalamic nuclei where expression persists in adults . Alternatively, employ genetic strategies to drive ectopic SPOCK3 expression in adult tissues of interest.

  • Challenge: Multiple SPOCK family members with potential redundancy
    Solution: Generate compound mutants (SPOCK1/SPOCK3, SPOCK2/SPOCK3 double knockouts) to address potential compensatory mechanisms. Use CRISPR-based screening to identify unique versus shared functions.

  • Challenge: Linking biochemical properties (MMP inhibition) to in vivo phenotypes
    Solution: Develop knock-in mouse models expressing SPOCK3 variants specifically deficient in MMP inhibition but retaining other functions. This would help attribute specific phenotypes to particular molecular functions.

  • Challenge: Translating mouse findings to human relevance
    Solution: Perform comparative studies examining SPOCK3 expression and function in human brain tissue samples or organoids. Consider screening for SPOCK3 variants in human neurodevelopmental disorder cohorts.

Addressing these challenges through the proposed methodological innovations would significantly advance understanding of SPOCK3's functions in neurodevelopment and potentially identify translational applications.

Product Science Overview

Introduction

SPARC (Secreted Protein Acidic and Rich in Cysteine), also known as Osteonectin or BM-40, is a matricellular protein that plays a crucial role in bone biology. It is one of the most abundant non-collagenous proteins in bone and is involved in various cellular processes, including cell proliferation, differentiation, and matrix assembly .

Structure and Domains

SPARC consists of three distinct domains:

  1. EF-hand domain: This domain is involved in calcium binding.
  2. Follistatin-like domain: This domain has unique properties and independent activity.
  3. Kazal-like domain: This domain also has unique properties and independent activity .
Function and Mechanism

SPARC is known for its regulatory role in extracellular matrix (ECM) mineralization, which is a key step in bone repair and remodeling. It affects the differentiation, maturation, and mineralization of osteoblasts, the cells responsible for bone formation . SPARC has a high affinity for type 1 collagen and hydroxylapatite (HA), which are essential components of the bone matrix .

Role in Osteoblasts

Studies have shown that SPARC bidirectionally regulates osteoblast mineralization in a dose-dependent manner. It influences the expression of various genes and proteins involved in bone formation, such as bone sialoprotein (BSP), osteocalcin (OCN), osteopontin (OPN), and alkaline phosphatase (ALP) . Additionally, SPARC interacts with collagen and activates the P38 pathway, which is crucial for osteoblast function .

Recombinant SPARC/Osteonectin 3 in Research

Recombinant SPARC/Osteonectin 3 is used in research to study its effects on bone biology and its potential therapeutic applications. By using recombinant proteins, researchers can investigate the specific roles and mechanisms of SPARC in various cellular processes and disease models .

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.