OSM Mouse

Oncostatin-M Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Receptor Interactions and Species Specificity

Mouse OSM exhibits distinct receptor-binding properties compared to human OSM (hOSM):

FeatureHuman OSMMouse OSM
Receptor ComplexesType I (LIFR/gp130) and Type II (OSMRβ/gp130)Type II (OSMRβ/gp130) only
Cross-Species ActivityActivates murine LIFR/gp130No activation of human receptors
Signaling PathwaysSTAT1/STAT3STAT3-dominated
Evolutionary SpecializationDual-receptor activationOSMRβ-specific

This divergence arises from amino acid differences in the AB loop region, which dictate receptor specificity . mOSM binds murine OSMRβ with high affinity but exhibits low-affinity interaction with LIFR, resulting in limited STAT3-mediated signaling .

Hematopoietic Stem Cell (HSC) Regulation

  • OSMRβ-Dependent Signaling: In adult mouse HSCs, OSM induces 171 genes (e.g., cell motility, adhesion regulators) via OSMRβ. Knockout (Osmr Δ/Δ) abolishes 88% of these responses .

  • Aging Phenotypes: Elevated Osmr transcript levels correlate with HSC aging, implicating OSM in age-related hematopoietic dysfunction .

Inflammation and Disease Models

  • Keratinocyte Inhibition: mOSM suppresses proliferation and differentiation markers in keratinocytes, linked to psoriasis and atopic dermatitis .

  • Metastasis Promotion: OSM enhances premetastatic niche formation in murine breast cancer models, increasing circulating tumor cells and lung metastasis .

  • Adipose Dysfunction: Adipocyte-specific Osmr knockout mice show impaired lipid metabolism independent of obesity .

In Vivo Applications

  • Recombinant mOSM: Used to study liver LDL receptor upregulation, endothelial IL-6 production, and synovial fibroblast activation .

  • Antibody Tools: Monoclonal antibodies (e.g., MAB495) enable ELISA and Western blot detection without cross-reactivity to human OSM .

Key Research Findings

Recent studies highlight mOSM’s functional complexity:

Study FocusKey InsightSource
HSC Transcriptomics151/171 OSM-induced genes require OSMRβ; enriched in cell adhesion
Receptor EvolutionAB loop mutations drive receptor specialization in mOSM
Cancer MetastasisOSM synergizes with TGF-β1 to promote intravasation and lung spread
Inflammatory Bowel DiseaseElevated OSM correlates with active colitis in murine models

Comparative Insights Across Species

While hOSM retains cross-species activity (activating murine LIFR), mOSM cannot engage human receptors due to structural incompatibilities . This limits translational studies but provides a model for OSMRβ-specific effects. Evolutionary analysis suggests mOSM’s receptor specificity emerged after gene duplication events .

Product Specs

Introduction
Oncostatin M, a member of the cytokine family that also includes leukemia-inhibitory factor, granulocyte colony-stimulating factor, and interleukin 6, is a growth regulator. This cytokine inhibits the growth of various tumor cell lines and regulates cytokine production from endothelial cells, such as IL-6, G-CSF, and GM-CSF.
Description
Recombinant Mouse Oncostatin M, produced in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 20.4 kDa, comprising 181 amino acids. The purification process of OSM involves proprietary chromatographic techniques.
Physical Appearance
White, sterile-filtered lyophilized (freeze-dried) powder.
Formulation
The OSM protein was lyophilized from a 0.2 µm filtered solution concentrated in 1xPBS with a pH of 7.4.
Solubility
For reconstitution of lyophilized Oncostatin M, sterile 18 MΩ-cm H2O is recommended at a concentration of at least 100 µg/ml. This solution can be further diluted into other aqueous solutions.
Stability
Lyophilized Oncostatin M remains stable at room temperature for up to 3 weeks. However, it is recommended to store it desiccated below -18°C. Upon reconstitution, Oncostatin M should be stored at 4°C for 2-7 days. For long-term storage, it is advised to add a carrier protein, such as 0.1% HSA or BSA, and store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is greater than 95.0%, as determined by: (a) RP-HPLC analysis and (b) SDS-PAGE analysis.
Biological Activity
The ED₅₀, determined by the dose-dependent stimulation of NIH-3T3 mouse embryonic fibroblast cell proliferation, is less than 1 ng/ml. This corresponds to a specific activity greater than 1.0 x 10⁶ units/mg.
Synonyms
Oncostatin-M, OSM, OncoM.
Source
Escherichia Coli.
Amino Acid Sequence
NRGCSNSSSQ LLSQLQNQAN LTGNTESLLE PYIRLQNLNT PDLRAACTQH SVAFPSEDTL RQLSKPHFLS TVYTTLDRVL YQLDALRQKF LKTPAFPKLD SARHNILGIR NNVFCMARLL NHSLEIPEPT QTDSGASRST TTPDVFNTKI GSCGFLWGYH RFMGSVGRVF REWDDGSTRS R.

Q&A

What is Oncostatin M (OSM) and how does it function in mouse models?

Oncostatin M is a member of the interleukin-6 (IL-6) family of cytokines that exhibits both pro-inflammatory and anti-inflammatory properties depending on cellular and disease contexts. In mouse models, OSM has been identified as a potent inducer of muscle atrophy, signaling primarily through the JAK/STAT3 pathway. Mouse OSM (mOSM) specifically signals through a heterodimeric receptor complex that incorporates the mOSM-specific receptor mOSMRβ, forming what is known as receptor complex II . This signaling pathway activates downstream targets including muscle atrophy-related genes such as Atrogin1. Mouse OSM has distinct biological effects compared to human OSM (hOSM) or bovine OSM (bOSM) when studied in mouse systems, which is a critical consideration for experimental design .

How does mouse OSM differ from human OSM in signaling mechanisms?

A crucial distinction for researchers to understand is that mouse OSM (mOSM) and human OSM (hOSM) utilize different receptor complexes when studied in mouse systems. In mouse cells, mOSM signals through a heterodimeric receptor complex incorporating the mOSM-specific receptor mOSMRβ, while hOSM and bovine OSM (bOSM) instead utilize the mouse LIF receptor (mLIFRβ) rather than mOSMRβ . This divergence in receptor utilization leads to significantly different biological outcomes. Mice overexpressing hOSM or bOSM exhibit phenotypes resembling leukemia inhibitory factor (LIF) activation, including hematologic abnormalities, weight loss, and severe osteosclerosis. In contrast, mice overexpressing mOSM show more modest changes without these specific pathologies . This receptor specificity means that studies using non-mouse OSM in mouse models may actually be reporting LIF biology rather than authentic OSM effects.

What mouse models are available for studying OSM signaling?

Several mouse models have been developed to investigate OSM signaling, each with specific characteristics:

  • Constitutive Overexpression Models: Mice engineered to stably overexpress mOSM, hOSM, or bOSM through retrovirus-mediated gene transfer allow comparison of different OSM variant effects .

  • Constitutive Knockout Models: The Osmr^tm1Mtan model disrupts the first coding exon of Osmr by knock-in of a lacZ and neomycin expression cassette, creating a non-conditional knockout. These mice are viable and fertile but show reduced numbers of erythroid and megakaryocytic progenitors .

  • Conditional Knockout Models: The B6;129-Osmr^tm1.1Nat/J model (Osmr^fl/fl) can be paired with tissue-specific Cre recombinase systems (like Mx1-Cre) to achieve conditional deletion of Osmr exon 2 .

  • Receptor Fusion Protein Models: Engineered OSM signaling inhibitors using receptor fusion proteins containing the ligand binding domains of murine OSMRβ and murine GP130 have been developed for targeted inhibition studies .

Each model system offers distinct advantages depending on the specific research question being addressed.

How can researchers effectively validate OSMR knockout efficiency in conditional mouse models?

Validating OSMR knockout efficiency requires a multi-level approach examining both mRNA and protein expression across relevant tissues. The research literature indicates significant variability in knockout efficiency depending on tissue type, even within the same model system. When using the B6;129-Osmr^tm1.1Nat/J conditional knockout model with Mx1-Cre, researchers should:

  • Verify genomic recombination: Use PCR to confirm the presence of loxP sites (typically resulting in a 230bp product compared to 170bp in wild-type) and Cre recombinase (100bp product) .

  • Assess mRNA expression: Quantify Osmr transcript levels using real-time PCR in multiple tissues. Important finding: Some models may show inconsistent reduction in mRNA levels across tissues despite confirmed genomic recombination .

  • Evaluate protein expression: Western blot analysis of OSMR protein is essential, as transcript reduction doesn't always correlate with protein reduction. In Osmr^fl/fl Mx1-Cre mice, spleen tissue showed complete loss of OSMR protein, while bone marrow, kidney, and lung had varying levels of reduction .

  • Functional validation: Test OSM-induced gene expression changes through RNA-seq analysis of target cells. Even with partial OSMR reduction, a subset of OSM-responsive genes may show altered induction patterns. Specifically, in hematopoietic stem cells from Osmr^fl/fl Mx1-Cre mice, 17 genes were no longer induced by OSM stimulation while others remained responsive .

This comprehensive validation approach is necessary because the commonly used B6;129-Osmr^tm1.1Nat/J model has shown variable and tissue-dependent impacts on mRNA and protein expression, which directly affects experimental interpretation.

What methodological approaches should be used to measure mouse OSM levels in experimental samples?

Accurate quantification of mouse OSM requires specific technical considerations:

  • ELISA-based detection: The mouse OSM DuoSet ELISA system employs a sandwich enzyme-linked immunosorbent assay design for quantification in cell culture supernatants, serum, and plasma. This method requires:

    • Generating a four-parameter logistic (4-PL) curve-fit from standard readings

    • Sample dilution optimization (with multiplication of concentration by the dilution factor)

    • Careful selection of reagent diluent components, particularly bovine serum albumin (BSA)

  • Sample preparation considerations:

    • For serum/plasma samples, reagent diluent optimization should begin with PBS supplemented with 10-50% animal serum

    • High-quality BSA is crucial for optimal performance, as impurities like proteases or soluble receptors can interfere with detection

    • Standard curve suppression may indicate issues with BSA quality

  • Specificity verification: The assay exhibits no cross-reactivity with related factors including mouse CT-1, gp130/Fc Chimera, IL-6, IL-11, LIF, LIF Rα, OSM Rβ/Fc Chimera, rat CNTF, or human OSM at concentrations up to 50 ng/mL .

These methodological details are essential for generating reliable measurements of mouse OSM in experimental contexts.

How does OSM contribute to cancer-associated muscle wasting in mice, and what experimental approaches best study this phenomenon?

OSM has been identified as a potent inducer of muscle atrophy in the context of cancer cachexia. Experimental approaches to study this mechanism should include:

  • In vitro myotube models: Primary myotubes treated with OSM exhibit cellular atrophy via the JAK/STAT3 pathway. This system allows for direct assessment of OSM's effects on muscle cells independent of other systemic factors .

  • Transcriptomic analysis: RNA sequencing of OSM-treated myotubes reveals induction of various muscle atrophy-related genes, including Atrogin1. This approach enables identification of the molecular signature of OSM-induced atrophy. Compared to other IL-6 family cytokines (IL-6, LIF), OSM produces more extensive transcriptomic changes in myotubes .

  • OSM overexpression models: Mice engineered to overexpress OSM develop muscle wasting, providing an in vivo system to study OSM-induced atrophy .

  • Muscle-specific OSMR knockout models: Mice with muscle-specific deletion of OSMR show resistance to tumor-driven muscle wasting, demonstrating the necessity of OSM signaling in this process .

  • Therapeutic neutralization studies: Administration of OSM-neutralizing antibodies preserves muscle mass and function in tumor-bearing mice, allowing assessment of intervention efficacy .

These complementary approaches provide a comprehensive framework for investigating OSM's role in cancer cachexia, from molecular mechanisms to potential therapeutic applications.

What control conditions are essential when designing experiments with OSM in mouse models?

Proper experimental design for OSM studies in mice requires several specific controls:

  • Species-matched controls: Due to the significant differences in receptor utilization between mouse and human OSM in mouse systems, experiments must use species-appropriate controls. Studies using mouse OSM should not be directly compared to those using human or bovine OSM without accounting for their different signaling pathways .

  • Genotype controls: For OSMR knockout studies, appropriate controls include:

    • Cre-only controls (e.g., Mx1-Cre without floxed Osmr) to account for Cre expression effects

    • Wild-type C57BL/6J mice as negative controls for genomic validation

    • Tissue-matched samples from control and knockout mice for protein expression analysis

  • Treatment controls for OSM stimulation experiments:

    • Vehicle-treated controls matching the carrier solution for recombinant OSM

    • Time-matched controls for kinetic studies of OSM response

    • Concentration gradients to establish dose-response relationships

  • Related cytokine controls: Include other IL-6 family cytokines (IL-6, LIF) as comparators to distinguish OSM-specific from family-general effects .

These control conditions are essential for proper interpretation of OSM-related phenotypes and mechanisms in mouse model systems.

How should researchers interpret contradictory findings between different OSM mouse models?

Contradictory findings between OSM mouse models can arise from several factors that require careful consideration:

  • Species-specific receptor binding differences: Human OSM binds to both type I (gp130/LIFR) and type II (gp130/OSMR) receptor complexes in human cells, but predominantly signals through type I receptors in mouse cells. This fundamental difference means that studies using human OSM in mouse models are essentially studying a different signaling pathway than those using mouse OSM .

  • Model-specific variation in knockout efficiency: The B6;129-Osmr^tm1.1Nat/J conditional knockout model shows tissue-dependent variability in OSMR reduction. Spleen tissue may show complete protein loss while bone marrow exhibits only partial reduction. This variability can lead to seemingly contradictory phenotypes depending on the tissue examined .

  • Cell-autonomous versus non-cell-autonomous effects: Some phenotypes in OSMR knockout mice may result from both direct cellular effects and indirect effects through altered signaling in other cell types, complicating interpretation .

  • Transcript versus protein discrepancies: RNA-seq analysis has revealed cases where Osmr transcript levels appear unchanged despite functional evidence of altered OSM signaling, indicating potential post-transcriptional regulation mechanisms .

When facing contradictory findings, researchers should:

  • Carefully assess the specific OSM variant used (mouse, human, or bovine)

  • Validate knockout efficiency at both mRNA and protein levels in relevant tissues

  • Consider using multiple complementary approaches (e.g., genetic knockout and antibody neutralization)

  • Employ "humanized" murine OSM variants or receptor fusion proteins for more translatable studies

What considerations should guide the selection of appropriate OSM detection methods in mouse tissues?

Selecting appropriate OSM detection methods requires consideration of several experimental factors:

  • Sample type optimization:

    • For serum and plasma samples, reagent diluent optimization should begin with PBS supplemented with 10-50% animal serum

    • Cell culture supernatants typically require less complex sample preparation but may contain lower OSM concentrations requiring sensitive detection methods

  • Assay specificity verification:

    • Confirm that the detection method distinguishes mouse OSM from related cytokines like IL-6 and LIF

    • Verify lack of cross-reactivity with receptor components (gp130, OSMRβ, LIFRα) that may be present in biological samples

  • Data analysis approaches:

    • For ELISA data, compare 4-parameter logistic (4-PL) curve fitting with alternatives like plotting mean absorbance against concentration

    • Consider logarithmic transformation of both OSM concentrations and optical density values for linearization when appropriate

  • Tissue-specific considerations:

    • For tissue with high protease activity, include appropriate protease inhibitors during sample preparation

    • Account for potential matrix effects by preparing standards in matched matrix when possible

  • Complementary approaches:

    • Combine protein detection methods (ELISA, Western blot) with mRNA analysis (qPCR, RNA-seq) for comprehensive assessment of OSM expression and signaling

    • Consider functional readouts of OSM activity (STAT3 phosphorylation, target gene induction) alongside direct OSM measurement

These considerations ensure selection of detection methods that provide accurate and interpretable data on OSM levels in experimental systems.

How can researchers distinguish between OSM-specific and general IL-6 family effects in mouse studies?

Distinguishing OSM-specific effects from general IL-6 family effects requires several methodological approaches:

  • Comparative cytokine stimulation: Directly compare responses to mouse OSM, IL-6, and LIF in parallel experiments using identical conditions. RNA sequencing of primary myotubes treated with these cytokines reveals that OSM produces more extensive transcriptomic changes than IL-6 or LIF, with distinct gene expression profiles .

  • Receptor-specific knockout models: Compare phenotypes between:

    • OSMRβ knockout mice (affecting only OSM type II receptor signaling)

    • LIFRβ knockout mice (affecting LIF and human OSM signaling in mice)

    • gp130 knockout mice (affecting all IL-6 family cytokine signaling)

  • Cross-species comparisons: Analyze differential responses to mouse, human, and bovine OSM in mouse systems. While human and bovine OSM produce phenotypes resembling LIF activation (anemia, neutrophilia, lymphopenia, weight loss, and osteosclerosis), mouse OSM induces distinct effects with more modest lymphoid organ restructuring and no hematologic changes or weight loss .

  • Differential gene expression analysis: Identify OSM-specific gene signatures through RNA-seq. In hematopoietic stem cells, a subset of genes is regulated specifically by OSM-OSMRβ signaling, while others remain responsive even with reduced OSMR expression, suggesting differential regulation mechanisms .

  • Pathway inhibition studies: Use JAK/STAT pathway inhibitors alongside OSM treatment to determine signaling dependency. While multiple IL-6 family cytokines signal through the JAK/STAT3 pathway, the specific patterns and kinetics of activation may differ .

These approaches provide complementary evidence to distinguish truly OSM-specific effects from broader IL-6 family signaling outcomes.

What are the limitations of current mouse models for translating OSM research to human disease applications?

Current mouse models for OSM research have several important limitations for translational applications:

  • Receptor-binding differences: The most fundamental limitation is that human OSM binds both type I (gp130/LIFR) and type II (gp130/OSMR) receptor complexes in human cells, but preferentially signals through type I receptors in mouse cells. This means mouse studies using human OSM primarily reflect LIF-like signaling rather than authentic OSM effects .

  • Variable knockout efficiency in conditional models: The B6;129-Osmr^tm1.1Nat/J conditional knockout model shows tissue-dependent variability in OSMR reduction, with complete protein loss in some tissues but only partial reduction in others. This inconsistency complicates interpretation of phenotypes and may lead to false negative results .

  • Phenotypic differences between species: Mice overexpressing mouse OSM exhibit different phenotypes than those reported in human diseases involving OSM dysregulation, limiting direct translation of findings .

  • Incomplete gene repertoire conservation: Not all OSM-responsive genes are conserved between mice and humans, and even conserved genes may show different regulation patterns or functional outcomes .

  • Technical challenges in measuring OSM: Current detection methods may have differing sensitivity and specificity between mouse and human samples, complicating cross-species comparisons .

To address these limitations, researchers have developed:

  • "Humanized" murine OSM variants with altered receptor binding specificity

  • Receptor fusion protein inhibitors containing ligand binding domains of murine OSMRβ and GP130

  • Transgenic animals expressing these constructs for more translatable in vivo studies

These innovations aim to bridge the gap between mouse models and human disease applications in OSM research.

How should RNA-seq data from OSM-stimulated mouse cells be analyzed to identify key regulatory pathways?

RNA-seq analysis of OSM-stimulated mouse cells requires specific analytical approaches to identify key regulatory pathways:

  • Stringent differential expression criteria: Apply robust statistical thresholds (e.g., p < 0.01 and log2(FC) > 3 or < −3) to identify significantly differentially expressed genes. Remove predicted genes and pseudogenes to focus on functionally relevant targets .

  • Comparative stimulus analysis: Compare gene expression changes between:

    • OSM-stimulated versus vehicle-treated control cells

    • OSM-stimulated wild-type versus OSM-stimulated OSMR-deficient cells

    • OSM versus other IL-6 family cytokine (IL-6, LIF) stimulation

  • Response pattern categorization: Classify OSM-responsive genes into distinct patterns:

    • Genes induced by OSM in control cells but not in OSMR-deficient cells (OSM-OSMR dependent)

    • Genes induced by OSM in both control and OSMR-deficient cells (OSMR-independent OSM responses)

    • Genes showing enhanced responses to OSM in OSMR-deficient cells (potential compensatory mechanisms)

  • Pathway enrichment analysis: Apply gene set enrichment analysis to identify biological processes associated with OSM-responsive genes. In hematopoietic stem cells, OSM-OSMR signaling enriches for signatures of epithelial-to-mesenchymal transition, endothelial cell proliferation, and ion channel binding .

  • Integration with protein-level data: Correlate transcriptomic changes with protein expression and phosphorylation status of key signaling molecules (especially JAK/STAT pathway components) to establish causal relationships .

This comprehensive analytical framework enables identification of authentic OSM-regulated pathways and distinguishes direct from indirect effects in mouse model systems.

What approaches can be used to therapeutically target OSM signaling in mouse disease models?

Several approaches have been developed to therapeutically target OSM signaling in mouse disease models:

  • Neutralizing antibodies: OSM-specific neutralizing antibodies have shown efficacy in preserving muscle mass and function in tumor-bearing mice, demonstrating their potential for treating cancer cachexia. This approach directly targets circulating OSM without affecting other IL-6 family cytokines .

  • Receptor fusion proteins: Engineered receptor fusion proteins containing the ligand binding domains of murine OSMRβ and murine GP130 act as potent and specific inhibitors of OSM signaling. These constructs can be delivered via viral vectors and have been successfully used in studies of hematopoietic stem cell niche interactions and irritable bowel disease .

  • Conditional genetic deletion: Tissue-specific OSMR knockout using Cre-loxP systems (e.g., muscle-specific OSMR deletion) prevents tumor-driven muscle wasting, providing proof-of-concept for tissue-targeted approaches .

  • JAK/STAT pathway inhibitors: Since OSM signals predominantly through the JAK/STAT3 pathway, inhibitors of this pathway can effectively block OSM-induced transcriptional changes. This approach affects multiple cytokine signaling pathways but may be useful when broader suppression is desired .

  • "Humanized" murine OSM variants: Modified murine OSM proteins with altered receptor binding specificity enable more accurate modeling of human OSM biology in mouse systems, facilitating translational therapeutic development .

These complementary approaches provide a toolkit for investigating OSM-targeted therapeutics across different disease contexts and with varying specificity profiles.

What emerging technologies might improve the study of OSM biology in mouse models?

Several emerging technologies hold promise for advancing OSM research in mouse models:

  • CRISPR-based precise genome editing: Beyond traditional knockout approaches, CRISPR technology enables:

    • Introduction of specific point mutations to dissect receptor binding domains

    • Knock-in of reporter constructs for live tracking of OSM expression and signaling

    • Generation of humanized mice expressing human OSM receptors for improved translational relevance

  • Single-cell RNA sequencing: This technology allows:

    • Identification of cell-specific responses to OSM stimulation

    • Characterization of heterogeneous effects within seemingly uniform cell populations

    • Mapping of OSM signaling networks across different cell types in complex tissues

  • Spatial transcriptomics: By preserving spatial information while assessing gene expression:

    • OSM production and response zones can be mapped within intact tissues

    • Local versus distant effects of OSM signaling can be distinguished

    • Microenvironmental influences on OSM biology can be characterized

  • Advanced receptor fusion proteins: Next-generation inhibitory constructs:

    • Can be engineered with improved pharmacokinetic properties

    • May include conditional activation domains for temporal control

    • Could incorporate tissue-targeting modules for site-specific effects

  • Multimodal in vivo imaging: Combining:

    • Reporter mice expressing fluorescent proteins under OSM-responsive promoters

    • Intravital microscopy for real-time visualization of signaling events

    • Whole-body imaging for tracking OSM-dependent processes longitudinally

These technologies promise to overcome current limitations in studying OSM biology and will likely yield more nuanced understanding of its roles in both physiological and pathological processes.

Product Science Overview

Discovery and Function

Oncostatin M was first identified for its ability to inhibit the growth of A375 melanoma cells and other human tumor cells, while not affecting the growth of normal human fibroblasts . It is primarily produced by activated T cells and macrophages . OSM plays a significant role in immune regulation, fibroblast proliferation, and inhibition of tumor cell proliferation .

Recombinant Mouse Oncostatin M

Recombinant Mouse Oncostatin M (mOSM) is a laboratory-produced version of the naturally occurring cytokine. It is typically derived from E. coli and has a molecular weight of approximately 21 kDa . The recombinant form is used extensively in research to study its biological activities and potential therapeutic applications.

Biological Activity

The biological activity of recombinant mOSM is measured using cell proliferation assays. For instance, it has been shown to induce the proliferation of NIH/3T3 mouse embryonic fibroblast cells . The effective dose (ED50) for this effect is typically in the range of 0.10-0.40 ng/mL .

Applications in Research

Recombinant mOSM is used in various research areas, including:

  • Cancer Research: Investigating its role in inhibiting tumor cell proliferation.
  • Immunology: Studying its effects on immune regulation and cytokine signaling pathways.
  • Cell Biology: Understanding its role in cell proliferation and differentiation.
Storage and Stability

Recombinant mOSM is typically lyophilized and can be reconstituted in sterile PBS. It is stable for up to 12 months when stored at -20 to -70°C in its lyophilized form . After reconstitution, it should be stored under sterile conditions at 2-8°C for up to one month or at -20 to -70°C for up to three 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 2024 Thebiotek. All Rights Reserved.