Osteocrin Human

Osteocrin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Bone Metabolism

  • Osteoblast Regulation: Enhances anabolic effects on osteoblasts, promoting bone formation during early differentiation . Expression decreases with osteoblast maturation and is suppressed by hydrocortisone and 1,25-dihydroxyvitamin D3 .

  • Therapeutic Potential: Inhibits mineralization in vitro, suggesting a role in modulating bone remodeling .

Cardiovascular and Renal Systems

  • Natriuretic Peptide (NP) Interaction: Binds NPR-C (natriuretic peptide receptor-C) with high affinity, antagonizing NP clearance and amplifying NP signaling .

  • Renoprotection: In ischemia–reperfusion injury models, OSTN overexpression reduced renal atrophy and fibrosis by 40–50% via suppression of the Wnt/β-catenin pathway .

  • Cardioprotection: Attenuates diabetic cardiomyopathy by enhancing proteasomal activity and reducing oxidative stress .

Renal Studies

  • Model: Ostn-knockout mice subjected to unilateral ischemia–reperfusion injury (IRI) .

  • Results:

    • Fibrosis Reduction: Renal fibrosis markers (TGF-β1, collagen I) decreased by 30% in OSTN-overexpressing mice.

    • Mechanism: OSTN inhibited Wnt/β-catenin signaling, reducing Mmp7 and Axin2 expression .

Cardiac Studies

  • Model: Streptozotocin-induced diabetic mice with cardiac-specific OSTN overexpression .

  • Results:

    • Functional Improvement: Ejection fraction increased by 25% compared to controls.

    • Molecular Effects: Enhanced proteasomal activity reduced ubiquitinated protein accumulation by 60% .

Study FocusModel/SystemKey OutcomeCitation
Renal ProtectionMurine IRI50% reduction in renal atrophy
Diabetic CardiomyopathySTZ-induced mice25% improvement in cardiac function
Osteoblast DynamicsHuman primary cells3.1-fold Ostn downregulation with HC

Clinical and Therapeutic Implications

  • Renal Disease: OSTN’s inhibition of fibrosis positions it as a candidate for acute kidney injury (AKI) to chronic kidney disease (CKD) transition therapy .

  • Bone Disorders: Potential use in osteoporosis by modulating osteoblast-osteoclast equilibrium .

  • Challenges: Pharmacokinetic optimization and targeted delivery remain hurdles due to its short half-life and broad receptor interactions .

Research Gaps and Future Directions

Product Specs

Introduction
Osteocrin is a secreted protein primarily found in bone tissue. Its expression peaks shortly after birth and declines with age. The protein is encoded by a 1280-bp mRNA and consists of 103 amino acids, with a molecular weight of 11.4 kDa. In osteoblastic cells, osteocrin production coincides with matrix formation and decreases as cells mature. Its expression is regulated by 1,25-dihydroxyvitamin D3, showing a dose-dependent decrease upon treatment. Osteocrin appears to function as a soluble regulator of osteoblasts, as prolonged exposure to osteocrin-conditioned media inhibited mineralization and reduced markers of osteoblast activity like osteocalcin and alkaline phosphatase expression.
Description
Recombinant Human Osteocrin is produced with a His Tag fused to its N-terminus. This His-Tagged Fusion Protein has a molecular weight of 13.6 kDa. It comprises 106 amino acid residues of the human Osteocrin protein and an additional 16 amino acid residues forming the His Tag.
Physical Appearance
The product is a white powder that has been filtered and lyophilized (freeze-dried).
Formulation
The product has been filtered through a 0.4 μm filter and lyophilized. The starting concentration prior to lyophilization was 0.5 mg/ml in a 0.05M Acetate buffer with a pH of 4.0.
Solubility
To create a working stock solution, add 0.1M Acetate buffer (pH 4.0) to the lyophilized powder to achieve a concentration of approximately 0.5 mg/mL. Ensure complete dissolution of the pellet. For dilutions requiring a higher pH, dilute the stock solution with the appropriate buffer to a concentration of 10 µg/ml. The antigen has limited solubility at higher concentrations. This product is not sterile. Filter it through a sterile filter before using it in cell culture.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize freeze-thaw cycles. Reconstituted protein remains stable at 4°C for a limited period, showing no significant change for up to two weeks.
Purity
Purity is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Osteocrin, Musclin, OSTN.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASHMVDVT TTEAFDSGVI DVQSTPTVRE EKSATDLTAK LLLLDELVSL ENDVIETKKK RSFSGFGSPL DRLSAGSVDH KGKQRKVVDH PKRRFGIPMD RIGRNRLSNS RG.

Q&A

What is Osteocrin and how was it initially characterized?

Osteocrin (OSTN, also known as Musclin) is a small secreted peptide (12-kD) originally identified in bone and muscle cDNA libraries. In humans, it is encoded by the OSTN gene (NM_198184, UniProt ID P61366) . Initial characterization demonstrated that OSTN is expressed in human skeletal tissue, particularly in osteoblasts in developing bone and at sites of bone remodeling. Methodologically, this was established through immunohistochemistry in developing human neonatal rib bone, showing intense immunoreactivity in osteoblasts on bone-forming surfaces, newly incorporated osteocytes, and some late hypertrophic chondrocytes . Further characterization involved time-dependent expression analysis in primary human osteoblast cultures, where OSTN expression decreased as cells differentiated, in contrast to alkaline phosphatase which increased during differentiation .

How does the molecular mechanism of Osteocrin differ from other secreted factors?

Osteocrin shares structural homology with natriuretic peptides but operates through a distinct mechanism. Unlike natriuretic peptides that directly activate their receptors, OSTN functions by binding with high affinity and specificity to the natriuretic peptide receptor C (NPR-C), a clearance receptor. By antagonizing NPR-C, OSTN prevents the clearance of natriuretic peptides, effectively increasing their local bioavailability and potentiating their signaling effects . This indirect mechanism allows OSTN to enhance cyclic guanosine monophosphate (cGMP) signaling in target tissues without directly triggering natriuretic activity. This mechanism has important implications for experimental design, as researchers must consider both direct OSTN effects and indirect effects through enhanced natriuretic peptide signaling.

In which human tissues is Osteocrin expressed and what methods best detect its expression?

Osteocrin exhibits tissue-specific expression patterns in humans:

TissueExpression LevelDetection Method
Neocortex (especially temporal and occipital)High (peaks during late-mid fetal stage)RNA-seq, FISH, RT-PCR
AmygdalaModerateRNA-seq
Parietal and frontal cortical regionsModerateRNA-seq
Skeletal tissue/osteoblastsHigh in developing bone and remodeling sitesImmunohistochemistry
Hippocampus, striatum, thalamus, cerebellumNot appreciableRNA-seq

For optimal detection, fluorescence in situ hybridization (FISH) has effectively demonstrated OSTN enrichment in the cortical plate of developing neocortex, while quantitative RT-PCR has confirmed induction in human fetal brain cultures. Researchers should note that OSTN expression shows developmental regulation, increasing during fetal development and peaking around the late-mid fetal stage, coincident with the onset of synaptogenesis in the cortical plate .

How has Osteocrin been evolutionarily repurposed in primates compared to other mammals?

Osteocrin represents a fascinating example of evolutionary repurposing in primates. While in mice and rats, Ostn is narrowly expressed in bone and muscle but not in the brain, in humans and other primates, OSTN has acquired novel expression in the neocortex. This evolutionary repurposing occurred through the acquisition of DNA regulatory elements within the OSTN gene locus. Methodologically, this was demonstrated through luciferase reporter gene expression studies using 2-kb genomic regions directly 5′ of the human OSTN and mouse Ostn transcriptional start sites. The −2kb hOSTN:Fluc construct was robustly induced in response to membrane depolarization in both mouse and human neurons, whereas the homologous mouse sequence did not drive substantial luciferase expression in neurons of either species .

Further analysis through truncations and deletions of the human OSTN regulatory region identified an 85-bp sequence located about 600 bp upstream of the transcription start site that is required for efficient activity-dependent induction of OSTN. This element contains tandem MEF2-binding sites that are specific to the human sequence and not present in the mouse homolog. Chromatin immunoprecipitation sequencing (ChIP-seq) confirmed MEF2 binding at this enhancer region in human but not mouse neurons . This evolutionary acquisition of MEF2-binding sites explains, at least in part, the human-specific neuronal expression of OSTN.

What are the methodological approaches to study cross-species differences in Osteocrin function?

To study cross-species differences in Osteocrin function effectively, researchers should employ a multi-faceted approach:

  • Comparative genomics: Analysis of the OSTN locus across species to identify conserved and divergent regulatory elements. This should include examination of enhancer elements, particularly the 85-bp enhancer containing MEF2-binding sites that is critical for human neuronal expression.

  • Transcriptional profiling: RNA-seq and quantitative RT-PCR of OSTN expression across tissues and developmental stages in multiple species. In research contexts, membrane depolarization experiments (using KCl or glutamate receptor agonists like NMDA) can be used to compare activity-dependent induction across species.

  • Reporter gene assays: Comparing the transcriptional activity of OSTN regulatory regions from different species using luciferase reporter constructs transfected into neurons or other relevant cell types, as demonstrated in the research where −2kb hOSTN:Fluc was induced in both mouse and human neurons, while −2kb mOstn:Fluc was not .

  • Functional studies in cross-species contexts: Expressing human OSTN in mouse cells or tissues to assess phenotypic effects, or conversely, knocking out OSTN in human cells to evaluate the consequences for neuronal development and function.

  • Epigenetic profiling: ChIP-seq for transcription factors (like MEF2) and histone modifications (such as H3K27ac) to map active regulatory elements in the OSTN locus across species.

How does Osteocrin regulate activity-dependent dendritic growth in human neurons?

Osteocrin has been demonstrated to restrict activity-dependent dendritic growth specifically in human neurons, representing a primate-specific mechanism for neuronal plasticity regulation. The molecular pathway involves calcium influx through L-type voltage-sensitive calcium channels or activation of NMDA receptors, leading to OSTN gene induction (>100-fold) in human neurons . OSTN is then secreted and acts in an autocrine or paracrine manner.

The specificity of this pathway to humans is explained by the evolutionary acquisition of MEF2-binding sites in the OSTN regulatory region, which enables activity-dependent transcription in response to neuronal depolarization. Methodologically, researchers investigating this pathway should:

  • Combine calcium imaging with real-time OSTN expression monitoring

  • Employ pharmacological manipulation of calcium channels and NMDA receptors

  • Use morphological analysis of neuronal dendrites under conditions of OSTN addition or depletion

  • Consider the temporal dynamics of OSTN expression, as it peaks during the late-mid fetal stage concurrent with synaptogenesis

While the exact downstream mechanisms remain to be fully elucidated, the data suggests OSTN may function as a brake on excessive activity-dependent growth, potentially contributing to the distinctive morphological and functional properties of human neurons .

What is the temporal and spatial expression pattern of Osteocrin in the developing human brain?

The temporal and spatial expression pattern of Osteocrin in the developing human brain reveals highly specific regulation:

Developmental StageBrain RegionOSTN ExpressionMethod of Detection
Early fetalNeocortexLow/beginningRNA-seq (BrainSpan Project)
Mid-fetalNeocortexIncreasingRNA-seq
Late-mid fetalNeocortex (cortical plate)Peak expressionRNA-seq, FISH
PostnatalNeocortexMaintainedRNA-seq
All stagesHippocampus, striatum, thalamus, cerebellumNot appreciableRNA-seq

Fluorescence in situ hybridization (FISH) analysis of human neocortex at postconception week 16 has confirmed that OSTN is specifically enriched in the cortical plate of developing neocortex. Within the neocortex, OSTN expression is particularly enriched in regions of temporal and occipital cortex, with lesser expression in parietal and frontal cortical regions .

Notably, this expression pattern differs from that of other activity-regulated secreted factors like BDNF. While BDNF is expressed in multiple human brain regions, OSTN expression is restricted primarily to the neocortex and to a lesser extent the amygdala. This suggests specialized roles for OSTN in higher cortical function development that are unique to primates .

How does Osteocrin expression correlate with osteoblast differentiation and activity?

Osteocrin expression exhibits a distinctive pattern in relation to osteoblast differentiation, providing a marker for osteoblast lineage cells that correlates with their activity state. In human cultured primary osteoblasts, OSTN expression decreases in a time-dependent manner as cells differentiate over 2, 3, and 6 days (p<0.02). This pattern contrasts with alkaline phosphatase, a classic marker of osteoblast differentiation, which increases with osteoblast maturation (p<0.05) .

Hormonal regulation studies have revealed that OSTN expression is modulated by key bone-active hormones:

HormoneEffect on OSTN ExpressionSignificance
Hydrocortisone (200 nM)Decreased by 1.5-, 2.3-, and 3.1-fold at 2, 3, and 6 days (p<0.05)Suggests suppression during glucocorticoid-induced bone loss
Low-dose estradiolDecreased time-dependently (p<0.05)Potential role in estrogen-regulated bone metabolism
High-dose estradiolNo significant changeIndicates dose-dependent effects

Spatially, OSTN immunoreactivity is most intense in active osteoblasts on bone-forming surfaces, newly incorporated osteocytes, and some late hypertrophic chondrocytes in developing human neonatal bone. In adult bone, expression is specifically localized to osteoblasts and young osteocytes at bone-forming sites . This expression pattern suggests OSTN functions primarily in active bone formation rather than bone maintenance or resorption.

These findings indicate that OSTN serves as a marker of osteoblast activity rather than differentiation state per se, with potential implications for understanding bone development and remodeling processes.

What experimental approaches effectively measure Osteocrin function in bone tissue?

Investigating Osteocrin function in bone tissue requires multiple complementary approaches:

  • Immunohistochemistry and in situ hybridization: These techniques allow spatial localization of OSTN protein and mRNA within bone tissue, respectively. They have successfully demonstrated OSTN expression in osteoblasts on bone-forming surfaces, newly incorporated osteocytes, and some late hypertrophic chondrocytes .

  • Primary osteoblast cultures: Time-course studies of OSTN expression in human primary osteoblasts under different conditions (e.g., with/without differentiation factors, hormones) provide insights into regulation. qRT-PCR can quantify expression changes .

  • Genetic manipulation models:

    • Loss-of-function: Ostn-knockout mice provide a model to study systemic effects of OSTN deficiency

    • Gain-of-function: Tissue-specific Ostn-overexpressing transgenic models allow assessment of ectopic or enhanced OSTN expression

  • Signaling pathway analysis: Since OSTN interacts with the natriuretic peptide system, experiments should measure cGMP levels and natriuretic peptide receptor expression/activity in bone cells with and without OSTN.

  • Functional assays for bone formation:

    • Alkaline phosphatase activity

    • Mineralization assays (Alizarin red staining)

    • Bone histomorphometry in animal models

    • Micro-CT analysis of bone microarchitecture

  • Co-culture systems: Osteoblast-osteoclast co-cultures can assess OSTN's effects on bone remodeling balance.

When designing these experiments, researchers should consider the temporal expression pattern of OSTN and its apparent correlation with active bone formation rather than mature osteoblast function.

What is the mechanism by which Osteocrin exerts renoprotective effects?

Osteocrin exerts renoprotective effects through multiple mechanisms centered on potentiation of the natriuretic peptide (NP) system and subsequent modulation of the Wnt/β-catenin signaling pathway. Research using Ostn-knockout (KO) mice and liver-specific Ostn-overexpressing transgenic mice crossed with KO (KO-Tg) has elucidated this complex mechanism .

The primary mechanism involves OSTN binding to natriuretic peptide receptor C (NPR-C) with high affinity, antagonizing NP clearance and increasing local NP availability. This leads to enhanced activation of natriuretic peptide signaling through NPR-A and NPR-B, resulting in increased cyclic guanosine monophosphate (cGMP) production. The elevated cGMP levels then inhibit the Wnt/β-catenin pathway, which is critically involved in renal fibrosis development .

This mechanism was demonstrated in proximal tubular cells (NRK52E), where OSTN significantly potentiated the inhibitory effects of natriuretic peptides on transforming growth factor β1–induced activation of the Wnt/β-catenin pathway. This effect was reproduced by a cGMP analog, confirming the role of this second messenger in mediating OSTN's effects .

In a unilateral ischemia–reperfusion injury (IRI) model, KO-Tg mice showed:

  • Significantly ameliorated renal atrophy

  • Reduced renal fibrosis and tubular injury

  • Decreased expression of fibrosis- and inflammation-related genes

  • Attenuated activation of the Wnt/β-catenin pathway and its downstream targets (Mmp7, Myc, and Axin2)

Importantly, while systemic Ostn-knockout mice showed only marginal worsening of renal fibrosis compared to wild-type mice after injury, ectopic overexpression of OSTN provided significant protection, suggesting therapeutic potential through administration rather than endogenous modulation .

How might Osteocrin's role in renal function influence experimental design for kidney disease models?

Osteocrin's renoprotective effects have significant implications for experimental design in kidney disease research:

  • Model selection considerations: Based on OSTN's demonstrated effectiveness in the acute kidney injury (AKI) to chronic kidney disease (CKD) transition model , researchers should prioritize models that capture this transition rather than focusing solely on either acute or chronic phases. The unilateral ischemia-reperfusion injury (IRI) model with extended follow-up (e.g., 21 days post-insult) provides an appropriate context for studying OSTN's effects.

  • Genetic manipulation approaches:

    • Simple knockout models may be insufficient, as Ostn-knockout mice showed only marginal worsening of renal fibrosis

    • Tissue-specific overexpression models (e.g., liver-specific like KO-Tg) better mimic therapeutic administration scenarios

    • Conditional knockout/overexpression can help distinguish developmental versus acute effects

  • Pathway analysis requirements: Comprehensive assessment of the Wnt/β-catenin pathway is essential, including:

    • PCR array analysis of pathway components

    • Measurement of downstream targets (Mmp7, Myc, Axin2)

    • Immunohistochemical localization of key proteins (e.g., MMP7, Wnt2) in corticomedullary proximal tubules

  • Cellular models: When using proximal tubular cells (e.g., NRK52E), researchers should incorporate:

    • Reporter plasmid transfection to monitor Wnt/β-catenin pathway activity

    • Combined treatments with OSTN and natriuretic peptides

    • cGMP analogs as positive controls

    • Transforming growth factor β1 as a fibrosis inducer

  • Outcome measurements: Beyond standard histological assessments of fibrosis and injury, researchers should evaluate:

    • Renal atrophy (kidney weight/body weight ratio)

    • Expression of fibrosis- and inflammation-related genes

    • Tubular injury markers

    • Wnt/β-catenin pathway activation status

  • Translational considerations: As OSTN potentiates natriuretic peptides locally with minimal systemic effects like hypotension (compared to direct NP administration), experiments should monitor both local kidney effects and systemic parameters to capture this advantage .

What are the optimal methods for quantifying Osteocrin expression in different tissue types?

Accurate quantification of Osteocrin expression requires tissue-specific methodological considerations. Based on established research protocols, the following approaches are recommended:

Tissue TypeRecommended MethodConsiderationsReference Standard
Neuronal tissueqRT-PCR with KCl/NMDA stimulationExpression is activity-dependent; membrane depolarization induces >100-fold expressionHuman fetal brain cultures as positive control
Bone tissueImmunohistochemistry + qRT-PCRSpatial localization is critical; expression varies with differentiation stateDeveloping bone as positive control
Kidney tissueqRT-PCR + PCR array for pathway analysisFocus on Wnt/β-catenin pathway componentsCommercially available qPCR template standards (NM_198184)
Cell culturesqRT-PCR with time-course analysisExpression changes over time with differentiation200 reactions per 1 nmol primer mix

For standardization across experiments, researchers should utilize:

  • Commercially available qPCR template standards (e.g., HK212445) containing exact quantities for transcript copy number calculation

  • Appropriate housekeeping genes that remain stable in the specific experimental condition

  • Calibration against known standards (50 × 10^7 copies, double-stranded DNA as provided in commercial kits)

For neuronal expression studies specifically, fluorescence in situ hybridization (FISH) has proven valuable for mapping expression patterns in developing human neocortex, allowing identification of specific neuronal subtypes expressing OSTN . This should be complemented with immunohistochemistry when studying protein localization.

How should researchers design experiments to distinguish direct Osteocrin effects from indirect effects via natriuretic peptide potentiation?

Designing experiments to distinguish direct Osteocrin effects from indirect effects mediated through natriuretic peptide (NP) potentiation requires careful methodological approaches:

  • Receptor blocking studies:

    • Include NPR-C antagonists to block OSTN binding to this receptor

    • Compare with NPR-A and NPR-B antagonists to block downstream NP signaling

    • The differential effects observed will help distinguish direct versus indirect mechanisms

  • Genetic models:

    • Use natriuretic peptide receptor knockout models (NPR-A, NPR-B, or NPR-C) to test OSTN effects in the absence of specific receptor signaling

    • Create dual knockout models (Ostn + natriuretic peptide genes) to assess potential synergistic or redundant effects

  • Signaling pathway isolation:

    • Directly measure cGMP levels as the key second messenger for natriuretic peptide signaling

    • Use cGMP analogs as positive controls to mimic NP effects

    • Compare direct OSTN application versus NP application across different concentrations

  • Cell-specific approaches:

    • Study cells that express different complements of natriuretic peptide receptors

    • Use siRNA knockdown of specific receptors to create cellular models with defined receptor expression

  • Temporal dynamics analysis:

    • Examine rapid signaling events (seconds to minutes) which are more likely direct effects

    • Compare with delayed responses (hours) which may involve transcriptional regulation and be indirect

  • Concentration-response relationships:

    • Compare dose-response curves for OSTN alone, NPs alone, and combinations

    • Non-additive effects at specific concentrations may indicate shared pathways

The experimental design successfully used in kidney cells demonstrates this approach, where transforming growth factor β1–induced activation of the Wnt/β-catenin pathway was inhibited by OSTN potentiating the effects of NPs, and this was reproduced by a cGMP analog . This systematic approach confirmed the mechanism operated through enhancement of NP signaling rather than a direct effect of OSTN.

What are the implications of Osteocrin's role in the acute kidney injury to chronic kidney disease transition for therapeutic development?

Osteocrin's demonstrated renoprotective effects in the acute kidney injury (AKI) to chronic kidney disease (CKD) transition have several significant implications for therapeutic development:

  • Novel therapeutic target: OSTN represents a potential therapeutic strategy against AKI-CKD progression, addressing a critical unmet need in nephrology. The effectiveness of ectopic OSTN overexpression in ameliorating renal atrophy, fibrosis, and tubular injury in mouse models provides proof-of-concept for therapeutic development .

  • Mechanistic advantages: OSTN offers a unique mechanism of action through potentiation of natriuretic peptide signaling by antagonizing NPR-C. This approach provides several advantages:

    • Local potentiation of NP action with minimal systemic effects such as hypotension

    • Enhanced cGMP signaling, which suppresses the Wnt/β-catenin pathway implicated in renal fibrosis

    • Similar mechanism to clinically used angiotensin receptor-neprilysin inhibitors, but with potentially greater tissue specificity

  • Biomarker potential: The association between OSTN gene polymorphisms and renal function decline identified in genome-wide association studies suggests potential for OSTN-related biomarkers to identify patients at risk for rapid CKD progression .

  • Therapeutic modalities:

    • Recombinant OSTN protein administration

    • Gene therapy approaches for tissue-specific OSTN expression

    • Small molecule mimetics that reproduce OSTN's NPR-C antagonism

    • Combination approaches with existing nephroprotective agents

  • Target patient populations: Based on experimental data, patients most likely to benefit would include:

    • Those recovering from AKI with risk factors for progression to CKD

    • Patients with conditions characterized by Wnt/β-catenin pathway activation in the kidney

    • Individuals with genetic variants in the OSTN gene associated with faster renal function decline

The renoprotective effects demonstrated in preclinical models suggest that OSTN-based therapeutics could potentially modify the disease course in the AKI-CKD transition, rather than simply providing symptomatic relief, representing an important advance in this field .

How might the primate-specific neuronal expression of Osteocrin influence translational neuroscience research?

The primate-specific neuronal expression of Osteocrin has profound implications for translational neuroscience research, challenging conventional approaches and opening new avenues of investigation:

  • Limitations of rodent models: The discovery that OSTN is expressed in primate but not rodent neurons due to evolutionary acquisition of MEF2-binding sites highlights a fundamental limitation of rodent models for studying certain aspects of human brain development and function . This finding suggests:

    • Critical evaluation of whether rodent models adequately recapitulate human neuronal development

    • Possible existence of other primate-specific genes with similar evolutionary histories

    • Need for complementary approaches using primate or human cellular models

  • Implications for neuronal plasticity and connectivity: OSTN's role in restricting activity-dependent dendritic growth specifically in human neurons suggests it may contribute to distinctive features of human neural networks . Research approaches should:

    • Focus on comparative analysis of dendritic complexity across species

    • Investigate whether OSTN influences synaptic specificity or circuit refinement

    • Consider the implications for neurodevelopmental disorders affecting connectivity

  • Methodological considerations for translational research:

    • Prioritize human neuronal models (iPSC-derived neurons, brain organoids) to study OSTN function

    • Develop humanized mouse models expressing human OSTN regulatory elements

    • Employ non-human primate models for in vivo studies when ethical and feasible

  • Implications for neurological disorders:

    • Investigate OSTN expression in neurodevelopmental disorders with altered connectivity

    • Consider whether OSTN dysregulation contributes to human-specific aspects of certain neurological conditions

    • Explore OSTN as a potential therapeutic target for conditions involving aberrant activity-dependent plasticity

  • Broader relevance to human brain evolution:

    • Use OSTN as a model to identify other genes with human-specific neuronal expression

    • Investigate whether OSTN contributes to expanded cortical development in primates

    • Examine the role of activity-dependent transcription in human-specific brain evolution

The finding that OSTN has been evolutionarily repurposed in the primate brain highlights the importance of considering species differences in gene regulation and function when translating basic neuroscience findings to human applications . This may necessitate re-evaluation of certain drug targets and disease mechanisms established solely in rodent models.

Understanding the Multifaceted Roles of Human Osteocrin in Research

Osteocrin represents a fascinating molecule with diverse tissue-specific functions and evolutionary significance. For researchers, understanding its complex biology requires appreciation of both its conserved and species-specific aspects. The evolutionary repurposing of OSTN in the primate brain, its established roles in bone development, and its emerging importance in renal protection all highlight the need for specialized methodological approaches tailored to each tissue context.

Advanced research should focus on integrating findings across these diverse biological systems, potentially revealing common mechanistic themes such as the modulation of natriuretic peptide signaling. The unique features of human OSTN, particularly its primate-specific neuronal expression, underscore the importance of appropriate model selection in translational research and highlight potential limitations of rodent models for certain aspects of human biology.

Product Science Overview

Discovery and Nomenclature

Osteocrin was first identified as a novel myokine, a type of cytokine produced and released by muscle cells in response to muscular contractions. The name “Osteocrin” is derived from its role in bone (osteo) and its secretory nature (crin). It is also referred to as Musclin due to its expression in muscle tissues .

Structure and Expression

The human Osteocrin protein shares 77% and 78% amino acid sequence identity with the rat and mouse proteins, respectively . It is synthesized as an 11 kDa proprotein that undergoes proteolytic processing to produce a mature 5 kDa C-terminal peptide . The protein is primarily expressed in bone and muscle tissues, and its secretion is increased with exercise .

Functions

Osteocrin plays several crucial roles in the body:

  1. Bone Growth and Differentiation: Osteocrin modulates osteoblastic differentiation, which is essential for bone formation and growth .
  2. Glucose Metabolism: It functions as an autocrine and paracrine factor linked to glucose metabolism in skeletal muscle .
  3. Neuronal Function: Osteocrin has been shown to regulate neuronal function, contributing to overall physical endurance .
  4. Cardiovascular Health: Recent studies suggest that Osteocrin may prevent diabetic cardiomyopathy, a condition characterized by damage to the heart muscle in people with diabetes .
Recombinant Human Osteocrin

Recombinant human Osteocrin is produced using E. coli expression systems. The recombinant protein is typically tagged with a His-tag for purification purposes and is available in lyophilized form for research use . It is used in various research applications to study its biological functions and potential therapeutic benefits.

Storage and Stability

Lyophilized recombinant Osteocrin is stable for up to 12 months when stored at -20 to -80°C. Reconstituted protein solutions can be stored at 4-8°C for 2-7 days or at -20°C for up to 3 months .

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.