OSM Human, 209 a.a refers to recombinant human Oncostatin M containing 209 amino acids. This multifunctional cytokine belongs to the Interleukin-6 (IL-6) subfamily, which also includes IL-11, leukemia inhibitory factor (LIF), ciliary neurotropic factor, cardiotrophin-1, and novel neurotropin-1. In research contexts, it functions as a growth regulator that can both stimulate and inhibit cell proliferation depending on the target cells. OSM plays roles in regulating neurogenesis, osteogenesis, and hematopoiesis, making it an important target for studying various physiological and pathological processes .
In the human body, OSM is primarily produced by activated immune and inflammatory cells. Understanding these natural cellular sources is important when designing experiments to study physiological conditions where OSM might play a regulatory role. The primary sources include:
Activated T lymphocytes
Monocytes and macrophages
Neutrophils
Endothelial cells
This production pattern suggests OSM plays important roles in immune response coordination and inflammation resolution, which should be considered when designing experimental models.
OSM exhibits pleiotropic effects across multiple biological systems, including:
Growth Regulation:
Stimulates proliferation of fibroblasts, smooth muscle cells, megakaryocytes, and vascular endothelial cells
Inhibits proliferation of several tumor cell lines, including solid tissue tumors, lung cancer, melanoma, and breast cancer cells
Cytokine Regulation:
Regulates production of IL-6, G-CSF, and GM-CSF from endothelial cells
Coordinates cytokine network responses in inflammatory settings
Developmental Processes:
Other Functions:
Enhances expression of low-density lipoprotein receptor in hepatoma cells
Plays roles in inflammatory responses and acute phase protein induction
Proper storage and handling are critical for maintaining the biological activity of OSM. Researchers should follow these guidelines:
Storage Recommendations:
Form | Temperature | Duration | Notes |
---|---|---|---|
Lyophilized | -80°C | Up to 6 months | Keep desiccated |
Lyophilized | -18°C | Long-term storage | Keep desiccated |
Reconstituted | 4°C | 2-7 days | For immediate use |
Reconstituted | -20°C | Up to 3 months | Minimize freeze-thaw cycles |
Reconstituted | -18°C | Long-term | Add carrier protein (0.1% HSA or BSA) |
Handling Guidelines:
Avoid repeated freeze-thaw cycles as these significantly reduce biological activity
Allow lyophilized protein to equilibrate to room temperature before opening
Use sterile techniques when handling the reconstituted protein
Aliquot reconstituted protein into smaller volumes to minimize freeze-thaw cycles
Following these storage protocols will help ensure consistent experimental outcomes and reproducibility in OSM-related research.
For optimal reconstitution of lyophilized OSM Human, 209 a.a, follow this step-by-step protocol:
Allow the lyophilized protein to reach room temperature before opening the vial
Reconstitute in sterile water or PBS to a concentration of at least 100 μg/ml
Gently mix by swirling or rotating the vial until completely dissolved (avoid vigorous shaking or vortexing)
For further dilutions, the reconstituted protein can be diluted in buffers containing carrier proteins
For long-term storage of reconstituted protein, it is recommended to add a carrier protein (0.1% HSA or BSA)
The solution should be clear after reconstitution. Any particulates or cloudiness may indicate protein degradation or denaturation. If this occurs, the preparation should not be used for experiments as it may yield unreliable results.
The biological activity of OSM Human, 209 a.a can be quantitatively assessed through several established methods:
Cell Proliferation Assays:
Growth Inhibition Assays:
Using cancer cell lines such as A375 melanoma or MCF-7 breast cancer cells
Measuring decreased proliferation or cell cycle arrest after OSM treatment
Cytokine Induction Assays:
Signaling Pathway Activation:
Monitoring phosphorylation of downstream signaling molecules (JAK/STAT, MAPK)
Using Western blotting or phospho-specific flow cytometry
These assays provide complementary information about OSM activity and should be selected based on the specific research question being addressed.
Several established cell lines are particularly useful for studying OSM activity in different research contexts:
Cell Line | Origin | Application | Response to OSM |
---|---|---|---|
TF-1 | Human erythroleukemia | Proliferation assays | Growth stimulation; ED50 < 2-10 ng/ml |
A375 | Human melanoma | Growth inhibition studies | Decreased proliferation |
MCF-7 | Human breast cancer | Anti-proliferative effects | Growth inhibition |
HepG2 | Human hepatocellular carcinoma | Hepatic responses | LDL receptor upregulation |
HUVEC | Human umbilical vein endothelial cells | Cytokine induction | IL-6, G-CSF, GM-CSF production |
Human fibroblasts | Primary or immortalized | Growth stimulation studies | Proliferation; ECM production |
Smooth muscle cells | Primary | Proliferative responses | Growth stimulation |
When selecting a cell line, researchers should consider receptor expression profiles (gp130, OSMR-β, LIFR-β) and the specific OSM-induced response they aim to study, as OSM effects can vary significantly between different cell types .
OSM exhibits a unique receptor binding pattern that distinguishes it from other IL-6 family cytokines:
Receptor Complexes:
Binding Mechanism:
Species Specificity:
Understanding these receptor interactions is crucial for designing experiments targeting specific OSM signaling pathways and for interpreting results in different experimental systems. Researchers should consider receptor expression patterns when selecting cell types for OSM studies.
OSM activates multiple intracellular signaling pathways that mediate its diverse biological effects:
JAK/STAT Pathway:
Primary pathway activated by OSM
JAK1, JAK2, and TYK2 kinases are activated upon receptor dimerization
Leads to phosphorylation and activation of STAT1, STAT3, and STAT5
STAT3 activation is particularly important for many OSM-mediated responses
MAPK/ERK Pathway:
OSM activates the Ras-Raf-MEK-ERK cascade
ERK1/2 phosphorylation leads to activation of various transcription factors
Important for OSM-induced proliferative effects
PI3K/AKT Pathway:
OSM can activate PI3K, leading to AKT phosphorylation
Contributes to cell survival signals and metabolic regulation
These pathways can be experimentally monitored to assess OSM activity and to understand the mechanisms underlying specific cellular responses. The activation patterns vary depending on cell type, receptor expression levels, and cellular context, contributing to the pleiotropic effects of OSM .
OSM shares structural and functional characteristics with other IL-6 family cytokines, but also has unique properties that should be considered in experimental design:
Cytokine | Shared Properties with OSM | Distinguishing Features | Experimental Considerations |
---|---|---|---|
IL-6 | Uses gp130 for signaling; Activates JAK/STAT pathway | Requires IL-6R (unlike OSM); Lower potency in growth inhibition | Include IL-6R blocking antibodies to distinguish effects |
LIF | Shares LIFR-β/gp130 receptor complex; Similar effects on some cell types | Cannot utilize OSMR-β; Different potency in many systems | Use OSMR-β selective signaling readouts for differentiation |
CNTF | Uses gp130 in receptor complex | Requires CNTFR-α; Primarily acts on neural tissues | Tissue-specific effects can help distinguish from OSM |
IL-11 | Signals through gp130; Roles in hematopoiesis | Requires IL-11R; More restricted biological activities | Selective receptor blocking can differentiate responses |
When designing experiments involving multiple IL-6 family cytokines, researchers should:
Use receptor-specific blocking antibodies to delineate pathway contributions
Compare dose-response relationships across different cytokines
Consider cell-type specific receptor expression patterns
Include appropriate positive and negative controls for each cytokine
Species-specific considerations are crucial when planning cross-species experiments with OSM:
Receptor Specificity Differences:
Experimental Design Implications:
When using human OSM in mouse models, consider that it may not fully recapitulate the physiological effects of mouse OSM
For in vitro studies with mouse cells, human OSM may produce results that differ from mouse OSM
Dose-response relationships may vary between species due to different receptor affinities
Control Recommendations:
In cross-species experiments, include species-matched controls when possible
Consider comparative studies with both human and mouse OSM when using mouse models
Validate key findings in species-specific systems to ensure translational relevance
These considerations are essential for accurate interpretation of experimental results and for translating findings between species, particularly in preclinical studies aimed at therapeutic development .
Researchers may encounter several challenges when using OSM in experimental systems:
Loss of Activity:
Variable Cellular Responses:
Cause: Cell passage number, density, or culture conditions affecting receptor expression
Solution: Standardize cell culture conditions; use cells within defined passage ranges; check receptor expression levels
Low Sensitivity in Bioassays:
Cause: Reduced receptor expression in target cells; inhibitors in media
Solution: Verify receptor expression; optimize cell culture conditions; serum-starve cells before OSM stimulation
Endotoxin Contamination:
Addressing these common issues requires careful experimental planning and appropriate controls to ensure reliable and reproducible results in OSM research.
Properly designed dose-response experiments are essential for characterizing OSM activity:
Following these guidelines will help generate reliable and interpretable dose-response data for OSM activity in various experimental systems.
Several methodologies are available to study OSM-receptor interactions at different levels:
Binding Assays:
Surface Plasmon Resonance (SPR):
Measures real-time binding kinetics
Can determine association (kon) and dissociation (koff) rates
Calculates equilibrium dissociation constant (KD)
Radio-ligand Binding:
Uses 125I-labeled OSM
Scatchard analysis for receptor number and affinity
Competition assays for comparative binding studies
Receptor Expression Analysis:
Flow Cytometry:
Quantifies surface receptor levels on intact cells
Can be combined with signaling readouts (phospho-flow)
Immunoblotting:
Measures total receptor protein levels
Can detect receptor dimerization using non-reducing conditions
qRT-PCR:
Quantifies receptor mRNA expression
Useful for monitoring regulation of receptor gene expression
Functional Readouts:
These methods provide complementary information about OSM-receptor interactions and should be selected based on the specific aspects of receptor biology being investigated.
Developing effective OSM antagonists requires systematic approaches:
Target Selection:
Direct OSM neutralization (antibodies against OSM)
Receptor blocking (antibodies against OSMR-β or gp130)
Pathway inhibition (JAK/STAT inhibitors)
In Vitro Validation:
Binding Assays:
Confirm direct interaction with target
Determine binding affinity and specificity
Functional Assays:
Inhibition of OSM-induced STAT3 phosphorylation
Blockade of OSM-dependent cell proliferation
Prevention of OSM-induced gene expression
Specificity Testing:
Cross-reactivity with related cytokines (LIF, IL-6)
Effects on other signaling pathways
Cellular Models:
Test in multiple cell types with different receptor expression patterns
Evaluate dose-dependent inhibition
Assess duration of antagonistic effects
Controls and Standards:
Include established inhibitors as positive controls
Use irrelevant antibodies or compounds as negative controls
Develop quantitative readouts for standardization
These methodological approaches provide a framework for developing and validating OSM antagonists that can be valuable tools for investigating OSM function in experimental systems.
OSM is being studied in multiple aspects of cancer biology:
Tumor Growth Regulation:
OSM demonstrates growth inhibition in multiple cancer cell types including melanoma, lung, and breast cancer cells
Mechanistic studies show this growth inhibition often involves cell cycle arrest rather than apoptosis
Paradoxically, OSM can promote growth in other tumor types, suggesting context-dependent effects
Metastasis Research:
Recent studies investigate OSM's role in promoting epithelial-to-mesenchymal transition (EMT)
OSM affects cancer cell migration and invasion through matrix remodeling
Research examining OSM production in the tumor microenvironment by infiltrating immune cells
Experimental Approaches:
These research applications highlight OSM's complex and sometimes contradictory roles in cancer biology, emphasizing the importance of context-specific experimental design.
Several experimental models are employed to study OSM's role in inflammatory processes:
In Vitro Models:
Co-cultures of immune cells and tissue-specific cells (e.g., synoviocytes, chondrocytes)
Primary human cell cultures stimulated with recombinant OSM
Cell line models for specific inflammatory responses
Ex Vivo Approaches:
Tissue explant cultures from inflammatory disease patients
Comparative studies of normal vs. inflamed tissues with OSM treatment
Patient-derived cells with genetic or pharmacological OSM pathway modulation
In Vivo Models:
Transgenic mice with tissue-specific OSM overexpression
Inflammatory disease models with OSM neutralization or receptor blockade
Humanized mouse models for translational studies
Analytical Methods:
Multi-parameter flow cytometry for cellular responses
Multiplex cytokine analysis for inflammatory networks
Histological assessment of inflammatory infiltration and tissue damage
These models provide complementary approaches to understand OSM's role in different aspects of inflammation, from molecular mechanisms to tissue-level effects and potential therapeutic interventions .
OSM's contribution to tissue repair processes can be investigated through:
Liver Regeneration Models:
Partial hepatectomy with OSM administration or neutralization
In vitro studies of hepatocyte proliferation and differentiation
Analysis of OSM expression during liver regeneration phases
Wound Healing Systems:
In vitro scratch assays with keratinocytes or fibroblasts
Ex vivo human skin models for wound repair assessment
In vivo excisional or incisional wound models with OSM modulation
Bone Remodeling:
Osteoblast and osteoclast co-culture systems
Bone fracture healing models
Micro-CT analysis of bone formation after OSM treatment
Experimental Approaches:
Temporal profiling of OSM expression during repair processes
Cell-specific knockout or overexpression of OSM receptors
Combinatorial treatments with other growth factors or cytokines
These experimental systems allow researchers to dissect OSM's specific contributions to different aspects of tissue regeneration, potentially informing therapeutic applications for promoting tissue repair .
Investigating OSM signaling specificity requires sophisticated methodological approaches:
Receptor Engineering:
CRISPR-Cas9 modification of receptor expression
Domain swapping between OSMR-β and LIFR-β to identify specificity determinants
Chimeric receptors to dissect signaling pathway activation
Pathway Dissection:
Small molecule inhibitors targeting specific branches of OSM signaling
Phosphoproteomic analysis to identify OSM-specific signaling nodes
Temporal analysis of signaling dynamics after OSM stimulation
Transcriptional Profiling:
RNA-seq to identify OSM-specific gene signatures
Comparison with other IL-6 family cytokines
ChIP-seq to map STAT binding sites after OSM treatment
Computational Approaches:
Systems biology modeling of OSM signaling networks
Prediction of pathway cross-talk and feedback mechanisms
Integration of multi-omics data for comprehensive pathway analysis
These methodologies enable detailed investigation of how OSM activates specific signaling pathways and gene expression programs, distinct from related cytokines, providing insights into its unique biological functions .
The recombinant form of Oncostatin M is produced using Escherichia coli (E. coli) expression systems. This method ensures high yield and purity of the protein. The recombinant protein has a molecular weight of approximately 23.6 kDa . The amino acid sequence of Oncostatin M is as follows:
AAIGSCSKEY RVLLGQLQKQ TDLMQDTSRL LDPYIRIQGL DVPKLREHCR ERPGAFPSEE TLRGLGRRGF LQTLNATLGC VLHRLADLEQ RLPKAQDLER SGLNIEDLEK LQMARPNILG LRNNIYCMAQ LLDNSDTAEP TKAGRGASQP PTPTPASDAF QRKLEGCRFL HGYHRFMHSV GRVFSKWGES PNRSRRHSPH QALRKGVRR
Oncostatin M is involved in a variety of biological functions:
Due to its diverse biological activities, Oncostatin M has potential therapeutic applications: