LIF Mouse

Leukemia Inhibitory Factor Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to LIF Mouse (Leukemia Inhibitory Factor)

Leukemia Inhibitory Factor (LIF) is a 20 kDa pleiotropic cytokine belonging to the interleukin-6 (IL-6) family. Recombinant mouse LIF, produced in E. coli, is a glycosylated monomer containing 180–202 amino acids, depending on the source . It shares 78–79% sequence identity with human LIF but exhibits mouse-specific biological activities, particularly in embryonic stem cell (ESC) maintenance and immune modulation . LIF binds to a heterodimeric receptor complex (LIFRα/gp130), activating downstream JAK-STAT3 and MAPK pathways .

Embryo Implantation

LIF is essential for uterine receptivity in mice. It induces decidualization and supports blastocyst attachment via STAT3 activation . LIFRα is expressed in luminal epithelial cells, while gp130 is localized to glandular epithelium and myometrium .

Stem Cell Maintenance

Mouse LIF sustains pluripotency in ESC cultures by:

  • Suppressing differentiation markers (e.g., SSEA-4) while maintaining SSEA-1 and SOX2 expression .

  • Inhibiting embryoid body formation, preserving self-renewal capacity .

Immune and Stress Responses

LIF induces CRH (corticotropin-releasing hormone) and POMC expression in trophoblasts, linking placental stress signaling to fetal neurodevelopment . In cancer models, anti-LIF antibodies enhance Th-1/Th-2 differentiation and reduce tumor growth by neutralizing LIF’s immunosuppressive effects .

Product Formulations

SupplierConcentrationFormulationActivity
ReproCELL10 µg/mL (03-0011)PBS + 1% BSA1×10⁶ units/mL
ReproCELL100 µg/mL (03-0011-100)PBS + 1% BSA1×10⁷ units/mL
Cell Guidance Systems0.1 mg/mL (reconstituted)10 mM acetic acid + TFA1×10⁸ units/mg

Usage Guidelines

  • ESC Culture: 10 ng/mL LIF maintains pluripotency for up to 15 passages .

  • Tumor Models: Anti-LIF immunization delays tumor onset in 4T1 breast cancer models .

  • Quality Control: Reconstituted LIF is stable for 1 month at 4°C and 3 months at -80°C .

Cancer Immunotherapy

  • Anti-LIF Antibodies:

    • Balb/c Tumor Model: Increased Th-1/Th-2 cytokine expression (e.g., IFN-γ, IL-2) and reduced tumor incidence .

    • LIFR vs. LIF Targeting: LIFR immunization inhibits tumor growth in 75% of mice vs. 25% with LIF alone, due to cross-reactivity with other cytokines (e.g., oncostatin M) .

Reproductive Biology

  • Embryo Implantation: LIF-deficient mice fail to implant blastocysts, rescued by recombinant LIF injection .

  • Stress Signaling: LIF-induced CRH in trophoblasts may regulate fetal brain development via the hypothalamic-pituitary-adrenal axis .

Product Specs

Introduction
Leukemia Inhibitory Factor (LIF) is a protein that plays a crucial role in maintaining embryonic stem cells. It prevents these cells from spontaneously changing into other cell types. LIF also has other functions, including aiding in the development of nerve cells that use acetylcholine, regulating stem cell versatility, influencing bone and fat metabolism, stimulating the growth of certain cell lines, and boosting the production of megakaryocytes (cells that produce platelets). The LIF proteins in humans and mice share a 78% similarity in their amino acid sequence.
Description
Recombinant Murine Leukemia Inhibitory Factor (LIF) is a single-chain protein produced in E. coli bacteria. It is not glycosylated, meaning it lacks attached sugar molecules. It comprises 181 amino acids and has a molecular weight of 20 kDa. The purification process utilizes specialized chromatographic methods to ensure its purity.
Physical Appearance
Sterile white powder obtained by freeze-drying.
Formulation
The Leukemia Inhibitory Factor (LIF) was freeze-dried from a sterile solution with a concentration of 1mg/ml. The solution contained 20mM Phosphate buffer with a pH of 7.4 and 0.02% Tween-20.
Solubility
To reconstitute the freeze-dried Leukemia Inhibitory Factor (LIF), it is advised to dissolve it in sterile water to a minimum concentration of 100µg/ml. This solution can be further diluted using other aqueous solutions.
Stability
While the lyophilized (freeze-dried) Leukemia Inhibitory Factor (LIF) remains stable at room temperature for up to 3 weeks, it is best stored in a dry environment below -18°C. Once reconstituted, store Leukemia Inhibitory Factor (LIF) at 4°C for a maximum of 7 days. For extended storage, freeze at -18°C. The addition of a carrier protein such as HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeated freezing and thawing cycles.
Purity
The purity is confirmed to be higher than 95.0% through the following analyses: (a) RP-HPLC (Reverse Phase High-Performance Liquid Chromatography), and (b) SDS-PAGE (Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis).
Biological Activity
The biological activity of murine LIF is evaluated using the M1 cell differentiation assay. The activity is determined to be less than 0.01 ng/ml, which translates to a specific activity of 100,000,000 IU/mg. The definition of a standard of 50 units is the LIF concentration in 1.0 mL of cell culture medium that triggers the differentiation of 50% of M1 colonies.
Synonyms
CDF, HILDA, D-FACTOR, Differentiation- stimulating factor, Melanoma-derived LPL inhibitor, MLPLI, Emfilermin, Leukemia inhibitory factor, LIF, DIA.
Source
Escherichia Coli.
Amino Acid Sequence
MSPLPITPVNATCAIRHPCHGNLMNQIKNQLAQLNGSANALFISYYTAQGEPFP NNVEKLCAPNMTDFPSFHGNGTEKTKLVELYRMVAYLSASLTNITRDQKVLNP TAVSLQVKLNATIDVMRGLLSNVLCRLCNKYRVGHVDVPPVPDHSDKEAFQR KKLGCQLLGTYKQVISVVVQAF.

Q&A

What is mouse LIF and what is its primary function in biological research?

Mouse Leukemia Inhibitory Factor (LIF) is a pleiotropic cytokine belonging to the interleukin-6 family. It functions as a lymphoid factor that promotes long-term maintenance of pluripotency in mouse embryonic stem cells (ESCs) by suppressing spontaneous differentiation . Structurally, mouse LIF is a highly conserved secretory glycoprotein approximately 20 kDa in size, containing 202 amino acid residues . Beyond stem cell maintenance, LIF plays crucial roles in cholinergic neuron differentiation, bone and fat metabolism, mitogenesis of certain factor-dependent cell lines, and promotion of megakaryocyte production in vivo . In laboratory applications, recombinant mouse LIF is typically derived from E. coli expression systems, comprising amino acid residues Pro25-Phe203 of the native protein .

How does mouse LIF maintain pluripotency in embryonic stem cells?

Mouse LIF maintains pluripotency through binding to its heterodimeric receptor complex consisting of LIF receptor alpha (LIFR/CD118) and glycoprotein 130 (gp130) . This binding activates several signaling pathways, primarily the JAK/STAT3 pathway, which regulates the expression of pluripotency-associated transcription factors. Scientific data demonstrates that mouse ESCs cultured with recombinant mouse LIF (10 ng/mL) for extended periods (15 days with multiple passages) continue to express pluripotency markers such as SSEA-1 and SOX2, while lacking differentiation markers like SSEA-4 . The methodological approach involves supplementing culture media with LIF at concentrations of 10 ng/mL, which effectively blocks differentiation signals and maintains the self-renewal capacity of ESCs through activation of STAT3-dependent transcriptional networks .

What is the difference between carrier-free and BSA-containing recombinant mouse LIF?

Recombinant mouse LIF is available in two primary formulations: with Bovine Serum Albumin (BSA) as a carrier protein or in carrier-free (CF) form . The BSA-containing formulation enhances protein stability, increases shelf-life, and allows for more dilute storage concentrations . This formulation is typically recommended for cell or tissue culture applications and as ELISA standards. In contrast, carrier-free LIF lacks BSA and is recommended for applications where the presence of BSA might interfere with experimental outcomes . Both formulations are typically lyophilized from filtered PBS solutions and require reconstitution at 0.2-1 mg/mL in sterile PBS before use . For methodological considerations, researchers should select the appropriate formulation based on their specific experimental requirements, particularly considering potential interference from BSA in downstream applications.

What are the molecular mechanisms by which LIF signaling interacts with epigenetic regulators in stem cells?

LIF signaling interfaces with epigenetic regulation through multiple mechanisms, including interactions with chromatin modifiers like LSD1 (Lysine-specific demethylase 1) and CHD7 (Chromodomain helicase DNA binding protein 7) . Research indicates that LSD1 operates cooperatively with CHD7 to regulate differentiation of mouse embryonic stem cells in response to LIF signaling . Additionally, LSD1 exhibits a scaffolding function that controls DNA methylation patterns in ESCs, which influences the cells' responsiveness to LIF . Methodologically, investigating these interactions requires chromatin immunoprecipitation followed by sequencing (ChIP-seq) to map binding sites of epigenetic regulators in relation to LIF-responsive genes. Complementary approaches include RNA-seq after knockdown/knockout of specific epigenetic factors to determine their effects on LIF-dependent transcriptional networks, and co-immunoprecipitation studies to identify direct protein-protein interactions between LIF signaling components and chromatin modifiers.

How does LIF signaling differ between embryonic stem cells and cancer stem cells?

LIF signaling exhibits context-dependent functions between embryonic stem cells (ESCs) and cancer stem cells (CSCs). In ESCs, LIF primarily maintains pluripotency through STAT3 activation , while in cancer contexts, LIF can promote tumor growth and CSC maintenance through overlapping but distinct pathways . In breast cancer models, LIF and LIFR expression correlates with tumor progression, with immunization against these proteins showing potential therapeutic effects . Methodologically, comparative phosphoproteomic analysis between ESCs and CSCs after LIF stimulation reveals differential activation of downstream pathways. Research approaches should include parallel treatment of ESCs and CSCs derived from the same tissue origin (e.g., mammary tissue) with recombinant LIF, followed by analysis of transcriptional responses and functional outcomes. A key difference is that while LIF withdrawal from ESCs triggers differentiation, in CSCs it may lead to reduced self-renewal but not necessarily differentiation, suggesting additional regulatory mechanisms in the cancer context .

What is the composition of the LIF receptor complex and how does it transduce signals?

The LIF receptor complex consists of two subunits: a ligand-binding component called LIF receptor alpha (LIFR/CD118) and a signal-transducing component, glycoprotein 130 (gp130) . When LIF binds to LIFR, it triggers heterodimer formation with gp130, activating associated Janus kinases (JAKs) through transphosphorylation. This leads to phosphorylation of STAT3 transcription factors, which dimerize and translocate to the nucleus to regulate gene expression. Methodologically, researchers can investigate this signaling cascade using phospho-specific antibodies in Western blotting to detect activated signaling components after LIF stimulation. Additionally, proximity ligation assays can visualize the formation of the LIFR-gp130 complex in intact cells. It's noteworthy that gp130 also participates in receptor complexes for other cytokines including Oncostatin M, Cardiotrophin-1, CNTF, IL-6, IL-11, and IL-27, creating potential for signaling crosstalk . This overlap in signaling components necessitates careful experimental design with appropriate controls when studying specific LIF effects.

How do soluble forms of LIFR affect LIF bioavailability and function?

Soluble forms of mouse LIFR alpha can be generated through alternative splicing mechanisms . These soluble receptors function as decoys that bind free LIF in the extracellular environment, thereby reducing the bioavailability of LIF for cell surface receptors. Methodologically, researchers can assess the impact of soluble LIFR by conducting competitive binding assays using recombinant soluble LIFR and measuring the subsequent reduction in cellular responses to LIF. In experimental systems, the ratio of membrane-bound to soluble LIFR forms provides a regulatory mechanism that fine-tunes LIF signaling intensity. To study this phenomenon, researchers should employ ELISA techniques to quantify soluble LIFR in culture supernatants or biological fluids, followed by correlation analysis with LIF-dependent cellular responses. Additionally, expression plasmids encoding the soluble splice variant can be transfected into cells to experimentally manipulate the balance between LIF sequestration and signaling, providing insights into how this regulatory mechanism influences stem cell maintenance or cancer cell behavior.

What are the optimal storage and handling conditions for recombinant mouse LIF?

Recombinant mouse LIF requires specific storage and handling protocols to maintain biological activity. Upon receipt, LIF should be stored immediately according to manufacturer recommendations . For concentrated LIF solutions (100 μg/mL), storage at 4°C provides stability for up to 6 months from the date of receipt . Working solutions should be prepared by diluting LIF in sterile tissue culture media to convenient concentrations, then storing at 4°C for routine use . Critically, repeated freeze-thaw cycles must be avoided as they can substantially reduce biological activity . For lyophilized preparations, reconstitution should be performed at concentrations of 0.2-1 mg/mL in sterile PBS . A manual defrost freezer is recommended for long-term storage to prevent temperature fluctuations . When designing experiments with extended timeframes, researchers should prepare small aliquots of LIF rather than repeatedly accessing a single stock solution, thus minimizing potential activity loss from handling and temperature changes.

How can researchers verify the biological activity of mouse LIF in their experimental systems?

Verifying the biological activity of mouse LIF is essential before conducting experiments. Several methodological approaches can be employed:

  • Pluripotency Maintenance Assay: Culture mouse ESCs with the LIF preparation (10 ng/mL) for 15 days with multiple passages, then analyze pluripotency marker expression (SSEA-1 positive, SSEA-4 negative) by flow cytometry .

  • Immunofluorescence Analysis: Visualize pluripotency markers in LIF-treated ESCs using immunostaining for SSEA-1 (red) and SOX2 (green), with DAPI counterstaining for nuclei .

  • Cytokine Induction Assay: Measure IL-6 secretion in M1 mouse myeloid leukemia cells after LIF treatment, with expected ED50 values of 0.1-0.6 ng/mL for this biological effect .

  • Differentiation Inhibition Assay: Establish embryoid bodies in the presence or absence of LIF and quantify differentiation markers, expecting inhibition of differentiation in LIF-treated cultures .

For accurate assessment, parallel testing with a verified standard LIF preparation is recommended, along with appropriate negative controls (cells cultured without LIF) and positive controls (cells cultured with validated LIF).

What considerations should be made when transitioning between different sources or batches of mouse LIF?

When transitioning between different sources or batches of mouse LIF, researchers must consider several methodological factors to ensure experimental consistency:

  • Activity Normalization: Different manufacturers may use varying activity units or protein quantification methods. Perform side-by-side bioactivity assays with both old and new LIF batches using pluripotency maintenance or IL-6 induction endpoints to establish equivalent working concentrations.

  • Formulation Differences: Check whether the new LIF preparation contains carriers like BSA or is carrier-free, as this affects applications and dosing . If transitioning between formulations, validate that the change does not impact experimental outcomes through parallel testing.

  • Expression System Variations: Verify whether the recombinant LIF derives from E. coli or mammalian expression systems, as this affects glycosylation patterns which may influence biological activity in certain contexts .

  • Overlap Period: During transition, maintain cultures with both old and new LIF preparations in parallel for at least 2-3 passages to confirm comparable maintenance of stem cell characteristics before completely switching.

  • Documentation: Thoroughly document batch numbers, sources, and any observed differences in cell morphology or marker expression to facilitate troubleshooting if inconsistencies arise later.

How can mouse LIF be used in neural differentiation protocols for embryonic stem cells?

Mouse LIF plays nuanced roles in neural differentiation protocols for embryonic stem cells. While LIF generally suppresses differentiation, it can be strategically incorporated into staged protocols to enhance specific neural outcomes:

  • Initial Maintenance Phase: Use LIF (10 ng/mL) to expand the undifferentiated ESC population before initiating neural induction .

  • Neural Induction Phase: Remove LIF and add retinoic acid to trigger neural differentiation, as demonstrated in protocols for generating embryoid bodies with neural potential .

  • Specific Neural Lineage Support: Reintroduce LIF at later stages to support particular neural subtypes, such as cholinergic neurons, which show enhanced differentiation in response to LIF signaling .

  • miRNA Modulation: Consider coupling LIF withdrawal with miRNA manipulation, particularly miR-124, which promotes neuronal differentiation of inner ear neural stem cells as shown in research studies .

The methodological approach involves careful timing of LIF addition and withdrawal, combined with appropriate morphogens and growth factors depending on the specific neural lineage desired. Monitoring of both pluripotency markers (SOX2) and neural markers (PAX6, Nestin, β-III tubulin) throughout the differentiation timeline provides essential quality control for these protocols.

What is the role of LIF signaling in cancer stem cells and how can it be targeted therapeutically?

LIF signaling in cancer stem cells (CSCs) represents a potential therapeutic target due to its roles in promoting tumor growth and maintaining stem-like properties in cancer cells. Research demonstrates that LIF and its receptor (LIFR) are expressed in various tumor types, including breast cancer, where they contribute to CSC maintenance .

Immunization strategies targeting LIF and LIFR have shown promising results in inhibiting tumor growth in mouse mammary tumor models . In experimental studies, while tumors developed in all control mice, LIF immunization reduced tumor formation to 75% of animals, and LIFR immunization (alone or combined with LIF) more dramatically inhibited tumor growth to only 25% of mice . Additionally, immunized mice exhibited delayed tumor appearance compared to controls .

Methodologically, researchers investigating LIF targeting can employ:

  • Active Immunization: Using truncated recombinant LIF and LIFR proteins to generate neutralizing antibodies in vivo .

  • Neutralizing Antibodies: Developing or utilizing existing monoclonal antibodies against LIF or LIFR for passive immunotherapy approaches.

  • Small Molecule Inhibitors: Screening compounds that disrupt LIF-LIFR binding or downstream signaling components.

  • Genetic Approaches: Using siRNA or CRISPR/Cas9 to knock down LIF or LIFR expression in tumor models.

These strategies should incorporate appropriate controls and assessment of both tumor growth kinetics and CSC frequency through limiting dilution assays.

How does the expression of LIF and LIFR correlate with cancer progression and prognosis?

Expression of LIF and LIFR demonstrates significant correlations with cancer progression and prognosis across multiple tumor types. In breast cancer models, immunohistochemical analysis of tumor sections confirms the expression of both LIF and LIFR proteins within tumor tissues . This expression pattern is functionally relevant, as experimental immunization against these proteins inhibits tumor growth, suggesting their role in promoting cancer progression .

Research indicates that LIFR expression can induce a dormancy phenotype in breast cancer cells disseminated to the bone marrow, potentially contributing to metastatic latency before eventual recurrence . This finding highlights the context-dependent functions of LIF signaling in cancer biology, where it may promote primary tumor growth but regulate dormancy in metastatic settings.

Methodologically, researchers investigating these correlations should employ:

These approaches provide critical insights into how LIF signaling components may serve as biomarkers for prognosis and potential targets for therapeutic intervention.

What emerging technologies can enhance the study of LIF signaling in developmental and pathological contexts?

Several cutting-edge technologies are transforming our ability to study LIF signaling dynamics:

  • CRISPR-based Genetic Screens: Genome-wide or targeted CRISPR screens can identify novel components of the LIF signaling network and potential synthetic lethal interactions in cancer contexts. This approach allows systematic identification of genes that, when disrupted, sensitize cells to LIF withdrawal or enhance LIF-dependent effects.

  • Single-cell Multi-omics: Integration of single-cell transcriptomics, proteomics, and epigenomics provides unprecedented resolution of heterogeneous cellular responses to LIF signaling. This technology reveals how individual cells within a population respond differently to LIF, capturing the full spectrum of cellular states during differentiation or cancer progression.

  • Optogenetic and Chemogenetic Control: Engineered systems allowing precise temporal control of LIF signaling components enable dissection of acute versus sustained signaling effects. These approaches overcome limitations of conventional knockout or overexpression studies by providing dynamic regulation of signaling intensity.

  • Organoid Models: Three-dimensional organoid cultures provide more physiologically relevant contexts for studying LIF functions compared to traditional 2D cultures. When derived from normal or cancerous tissues, organoids can recapitulate developmental processes or disease characteristics, offering improved models for studying LIF's roles in tissue organization and cancer.

  • Computational Modeling: Machine learning approaches integrating multi-dimensional datasets can predict complex relationships between LIF signaling intensity, duration, and cellular outcomes, generating testable hypotheses about context-dependent functions.

Methodologically, researchers should consider combining these technologies for comprehensive understanding of LIF biology in complex systems.

How might differences between human and mouse LIF impact translational research?

Differences between human and mouse LIF create important considerations for translational research:

  • Sequence Homology: Mouse LIF shares approximately 78% amino acid sequence identity with human LIF . While this high conservation enables cross-species activity, the 22% difference may affect binding affinity to receptors and downstream signaling intensity.

  • Pluripotency Regulation: Mouse ESCs require LIF for maintaining pluripotency in conventional culture, whereas human ESCs depend on FGF2 signaling, reflecting significant species differences in pluripotency network regulation. This fundamental distinction means findings about LIF functions in mouse pluripotency cannot be directly extrapolated to human contexts.

  • Glycosylation Patterns: Native mouse and human LIF exhibit different glycosylation profiles, which may affect protein stability, receptor binding, and immunogenicity. While E. coli-derived recombinant proteins lack glycosylation entirely , this absence may impact certain biological functions compared to naturally produced LIF.

  • Experimental Methodology: For translational relevance, researchers should:

    • Validate key findings using both species' proteins

    • Consider humanized mouse models expressing human LIF receptors

    • Test on human cell lines or patient-derived xenografts when possible

    • Perform cross-species dose-response studies to establish equivalent effective concentrations

  • Antibody Specificity: Due to sequence differences, antibodies against mouse LIF may not recognize human LIF with equal affinity, necessitating species-specific validation of immunological reagents.

These species-specific considerations must inform experimental design and interpretation when translating mouse findings to human applications.

Product Science Overview

Discovery and Nomenclature

LIF was first identified in the late 1960s when researchers observed that conditioned medium could induce differentiation in murine myeloid leukemia cells . Over the years, LIF has been rediscovered multiple times in different contexts, leading to various names such as Differentiation-Inducing Factor (D-Factor), Differentiation-Stimulating Factor (DSF), and Human Interleukin for DA cells (HILDA) .

Structure and Function

LIF is a glycoprotein that functions by binding to its specific receptor, LIF receptor (LIFR), which forms a heterodimer with the gp130 signal transducing subunit . This interaction activates several intracellular signaling pathways, including the JAK/STAT, PI3K, and MAPK pathways . These pathways are essential for the cytokine’s diverse biological effects.

Role in Embryonic Stem Cells

In mice, LIF is a key factor that prevents embryonic stem cells (ESCs) from differentiating . It is often added to stem cell culture media to maintain the pluripotency of ESCs, allowing them to self-renew and proliferate without differentiating . This property makes LIF invaluable in stem cell research and regenerative medicine.

Recombinant Production

Recombinant mouse LIF is produced using various expression systems, including bacterial, yeast, and mammalian cells . The recombinant form retains the biological activity of the native protein and is widely used in research and therapeutic applications.

Clinical Implications

LIF has shown promise in various clinical applications, including the treatment of myeloid leukemia and thrombocytopenia . Its ability to influence a wide range of cell types, including neurons, adipocytes, osteoblasts, and gonadal cells, makes it a versatile therapeutic agent .

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.