LIF Human

Leukemia Inhibitory Factor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

FeatureDetailsSource
Gene Homology75% homology between human and murine LIF genes.
Protein Size19.7–20 kDa (unglycosylated), 32–62 kDa (glycosylated).
Amino Acid Sequence180 residues (mature), including three disulfide bonds (Cys34-Cys156, Cys40-Cys153, and a third linking helix D to the A-B linker).
IsoformsSecreted (LIF-D), extracellular matrix-associated (LIF-M), and intracellular (LIF-T), generated via alternative splicing.

LIF adopts a four-helix bundle structure (up-up-down-down configuration), stabilized by disulfide bonds and glycosylation, which influences its receptor binding and stability .

Receptor Interaction and Signaling Pathways

LIF binds to a heterodimeric receptor complex comprising:

  1. LIF Receptor (LIFR): A type I transmembrane protein with two cytokine-binding modules (CBMs).

  2. gp130: A shared signal-transducing subunit critical for IL-6 family cytokines .

This interaction activates downstream pathways:

  • JAK/STAT3: Promotes stem cell pluripotency and anti-apoptotic responses.

  • MAPK/ERK: Regulates cellular differentiation and survival.

  • PI3K/AKT: Modulates immune cell function and tissue repair .

Key Roles in Physiology

FunctionMechanismEvidence
Stem Cell MaintenanceInhibits differentiation of embryonic stem cells (ESCs) via STAT3 activation.Critical for maintaining mouse ESC pluripotency; partial effects in humans .
Tissue RegenerationEnhances intestinal stem cell (ISC) proliferation post-radiation injury.LIF-deficient mice show impaired epithelial regeneration and reduced survival .
Immune ModulationInduces Treg differentiation and suppresses pro-inflammatory cytokines.Elevated in rheumatoid arthritis and tuberculosis; neutralizing antibodies improve tumor immunity .

Context-Dependent Effects

  • Cancer: Promotes cancer stem cell survival in breast and glioma models but inhibits myeloid leukemia differentiation .

  • Neural Repair: Stimulates Schwann cell proliferation and neural precursor migration post-injury .

Stem Cell Culture

ParameterMouse ESCsHuman ESCs
LIF DependencyHigh (STAT3 activation)Low (primarily supports survival)
Common UseRecombinant LIF + inhibitors of FGF2/MAPKFeeder layers or bFGF + TGFβ/Activin A

Cancer Therapy

  • Therapeutic Target: Anti-LIF antibodies reduce breast cancer progression by enhancing anti-tumor immunity .

  • Tumor Microenvironment: LIF promotes fibroblast activation and metastasis in gliomas and melanomas .

Regenerative Medicine

ApplicationFindingsSource
Radiation InjuryrLIF administration rescues intestinal regeneration and prolongs survival in mice.
AngiogenesisLIF-overexpressing MSCs secrete pro-angiogenic factors (VEGF, MCP-1), enhancing vascularization.

Glycosylation and Stability

  • Glycosylation Variability: Affects receptor binding and half-life (e.g., 20–25 kDa unglycosylated vs. 37–63 kDa glycosylated) .

Species-Specific Effects

Human LIF shares 79% homology with mouse LIF but lacks the same potency in inhibiting ESC differentiation .

Future Directions

  1. Radioprotection: Clinical trials evaluating rLIF for mitigating radiation-induced intestinal damage.

  2. Fertility Treatments: Exploring LIF’s role in improving implantation rates during IVF .

  3. Cancer Immunotherapy: Investigating anti-LIF antibodies to modulate tumor microenvironments .

Product Specs

Introduction
Leukemia Inhibitory Factor (LIF) is a protein that plays a crucial role in maintaining embryonic stem cells and preventing them from differentiating. It also has various other functions, including influencing the development of certain nerve cells, regulating stem cell versatility, impacting bone and fat metabolism, stimulating the growth of specific cell types, and promoting the production of megakaryocytes (cells that produce platelets). The human and mouse versions of LIF share a 78% similarity in their amino acid sequence.
Description
Recombinant Human Leukemia Inhibitory Factor (LIF) is produced in E. coli bacteria. It is a single, non-glycosylated polypeptide chain consisting of 180 amino acids with a molecular weight of 19.7kDa. This LIF protein is purified using specialized chromatographic methods, resulting in a highly pure product.
Physical Appearance
White powder, sterilized by filtration and freeze-dried.
Formulation
The Leukemia Inhibitory Factor (LIF) is provided as a freeze-dried powder. It was initially dissolved in a sterile solution of phosphate-buffered saline (PBS) at a concentration of 1mg/ml and adjusted to a pH of 7.4 before freeze-drying.
Solubility
To reconstitute the freeze-dried Leukemia Inhibitory Factor (LIF), it is recommended to dissolve it in sterile water at a concentration of at least 100µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Stability
The freeze-dried Leukemia Inhibitory Factor (LIF) remains stable at room temperature for up to 3 weeks. However, it is recommended to store it in a dry environment below -18°C for long-term storage. Once reconstituted, LIF should be stored at 4°C for up to 7 days. For extended storage, it's advisable to freeze it below -18°C. Adding a carrier protein such as HSA or BSA at 0.1% concentration can further enhance its stability during storage. Avoid repeated freezing and thawing cycles.
Purity
The purity of Leukemia Inhibitory Factor (LIF) is greater than 98% as determined by two different analytical methods: High-Performance Liquid Chromatography (RP-HPLC) and Sodium Dodecyl Sulphate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).
Biological Activity
The biological activity of Leukemia Inhibitory Factor (LIF) was assessed using the M1 cell differentiation assay. The ED50, which represents the concentration of LIF required to achieve 50% of the maximum biological response, was found to be less than 0.01 ng/ml. This translates to a specific activity of 100,000,000 International Units (IU) per milligram of protein.
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
SPLPITPVNA TCAIRHPCHN NLMNQIRSQL AQLNGSANAL FILYYTAQGE PFPNNLDKLC GPNVTDFPPF HANGTEKAKL VELYRIVVYL GTSLGNITRD QKILNPSALS LHSKLNATAD ILRGLLSNVL CRLCSKYHVG HVDVTYGPDT SGKDVFQKKK LGCQLLGKYK QIIAVLAQAF.

Q&A

What is LIF and what are its primary functions in human systems?

Leukemia Inhibitory Factor (LIF) is a multifunctional cytokine consisting of 180 amino acids in its human recombinant form. Structurally, it belongs to the interleukin-6 family of cytokines and initiates signal transduction through binding to the LIF receptor (LIFR) and glycoprotein 130 (gp130) .

LIF functions across multiple biological systems:

  • Maintenance of pluripotency in stem cells

  • Regulation of differentiation in multiple cell lineages

  • Modulation of inflammatory responses

  • Involvement in bone and fat metabolism

  • Neuronal cell development and function

In experimental systems, LIF is primarily utilized to maintain stem cells in an undifferentiated state by activating STAT3 signaling pathways, which subsequently regulate pluripotency-associated transcription factors .

How do LIF-dependent human iPSCs differ from bFGF-dependent iPSCs?

LIF-dependent and bFGF-dependent human induced pluripotent stem cells (iPSCs) represent distinct pluripotent states with unique characteristics:

CharacteristicLIF-dependent iPSCsbFGF-dependent iPSCs
Growth factor dependencyLeukemia Inhibitory FactorBasic Fibroblast Growth Factor
Pluripotency stateTypically considered more "naïve""Primed" pluripotent state
Colony morphologyMore dome-shaped coloniesFlat colony morphology
Single-cell dissociationBetter survivalTend to enter apoptosis
X-chromosome inactivationVariable, can maintain two active X chromosomesOne X chromosome usually inactive
Gene expression profileHigher expression of naïve markersHigher expression of primed markers

Importantly, research has demonstrated that LIF-dependent human iPSCs can be established without chemical inhibitors or sustained transgene expression when using transcriptionally enhanced OCT4 (M³O). This finding challenges the previous assumption that cytokine requirements define stem cell phenotypes, suggesting that bFGF and LIF signaling pathways may converge on common OCT4 target genes .

What experimental approaches are used to evaluate LIF signaling in cellular systems?

Methodologically sound evaluation of LIF signaling requires multiple complementary approaches:

  • Phosphorylation Analysis: Western blotting to detect phosphorylated STAT3 (pSTAT3), a key downstream effector of LIF signaling, typically at Tyr705 and Ser727 residues.

  • Transcriptional Activation Assays: Utilizing luciferase reporters driven by STAT3-responsive elements to quantify LIF-induced transcriptional activity.

  • Gene Expression Analysis: RT-qPCR assessing expression levels of LIF-responsive genes, including OCT4, SOX2, NANOG, KLF4, REX1, and STELLA .

  • Flow Cytometry: Measuring surface expression of LIF receptor components (LIFR and gp130) to assess receptor availability.

  • Immunofluorescence Microscopy: Visualizing nuclear translocation of STAT3 following LIF stimulation.

When interpreting results, researchers should note that expression levels of pluripotency markers (OCT4, SOX2, NANOG, KLF4, REX1, STELLA, FGF5 and T) are often similar between different pluripotent stem cell types, with differences typically less than 1.5-fold .

How does transcriptionally enhanced OCT4 establish LIF-dependent human iPSCs without signal transduction inhibitors?

The establishment of LIF-dependent human iPSCs without chemical inhibitors or sustained transgene expression represents a significant methodological advancement. This is achieved using M³O, a fusion protein combining OCT4 with the powerful transactivation domain of the myogenic master transcription factor MYOD .

Mechanistic Pathway:

  • M³O significantly enhances transcriptional activation by OCT4 while maintaining target gene specificity

  • When combined with SOX2, KLF4, and c-MYC (M³O-SKM), it increases iPSC colony formation efficiency 10-50 fold compared to standard OSKM protocols

  • Time required for iPSC colony formation is reduced by approximately 50%

  • The enhanced transcriptional activity appears to bypass the need for signaling pathway inhibitors traditionally required for LIF-dependent human iPSC maintenance

This approach differs fundamentally from previous methods that relied on inhibitors against ERK1/2 and GSK3 pathways, as well as constitutive expression of transgenes like OCT4 plus KLF4, or KLF2 plus KLF4 .

What mechanisms explain the therapeutic potential of targeting LIF in cancer immunotherapy?

LIF has emerged as a strategic therapeutic target in cancer due to its role in macrophage-mediated immunosuppression. The underlying mechanisms involve complex cellular interactions:

  • Immunosuppressive Microenvironment: LIF contributes to creating an immunosuppressive tumor microenvironment by modulating macrophage phenotypes toward an M2-like state.

  • Therapeutic Antibody Development: High-affinity therapeutic antibodies that potently inhibit LIF signaling, such as MSC-1, have demonstrated efficacy in immune-competent animal models of cancer .

  • Pathway Inhibition: LIF inhibition affects multiple downstream signaling cascades including JAK/STAT, PI3K/AKT, and MAPK pathways, which collectively influence tumor progression.

The experimental validation of LIF as a therapeutic target requires comprehensive assessment in appropriate models:

  • In vitro: Evaluation of LIF inhibitors on macrophage polarization, tumor cell proliferation, and immune cell function

  • In vivo: Testing in immune-competent animal models to assess effects on tumor growth, metastasis, and immune infiltration

  • Biomarker Analysis: Identification of predictive markers for response to LIF-targeting therapies

These approaches collectively support the development of LIF inhibitors as potential immunomodulatory cancer therapeutics .

How can researchers accurately distinguish between direct and indirect effects of LIF in complex biological systems?

Distinguishing direct from indirect effects of LIF presents significant methodological challenges. Recommended experimental approaches include:

  • Conditional Knockout Systems: Using inducible Cre-loxP systems to delete LIF or its receptor in specific cell types or at specific timepoints.

  • Receptor Chimeras: Constructing chimeric receptors containing the extracellular domain of an orthogonal receptor and the intracellular domain of LIFR to isolate LIF signaling effects.

  • Phosphoproteomic Analysis: Time-course studies measuring protein phosphorylation events to identify primary versus secondary signaling responses.

  • Transcriptomic Analysis with Temporal Resolution: RNA-seq at multiple timepoints following LIF stimulation, with and without protein synthesis inhibitors (e.g., cycloheximide), to distinguish immediate-early gene responses from secondary transcriptional events.

  • Spatial Analysis: Techniques such as single-cell RNA-seq combined with spatial transcriptomics to map LIF effects across different cell populations in complex tissues.

These approaches help construct cause-effect relationships and distinguish primary LIF signaling from downstream cascades that may involve multiple intervening factors.

What are the optimal conditions for maintaining LIF activity in experimental systems?

Preserving LIF bioactivity requires careful attention to storage and handling conditions:

ParameterOptimal ConditionNotes
Storage Temperature-80°C for long-term; -20°C for intermediateAvoid repeated freeze-thaw cycles
Carrier Protein0.1% HSA or BSA recommendedStabilizes protein during storage
Buffer CompositionPhosphate buffered (pH 7.2-7.4)Maintains protein structure
Working Concentration10-50 ng/ml for stem cell maintenanceCell type dependent
Activity TestingSTAT3 phosphorylation assayConfirms functional activity

For experimental reproducibility, it is essential to use purified LIF with confirmed biological activity. The purity should be verified by RP-HPLC and SDS-PAGE analysis, with activity confirmed through bioassays measuring STAT3 phosphorylation or maintenance of pluripotency markers in stem cells .

How can researchers effectively integrate LIF expression data with disease-gene relationship analyses?

Integrating LIF expression data with disease-gene relationships requires sophisticated bioinformatic approaches:

  • Literature-Based Methods: The LIF method (Literature data and Impact Factor) provides a framework for inferring disease-gene relationships by analyzing published literature and impact factors .

  • Network Analysis:

    • Construct disease-related gene networks using literature and public databases

    • Identify disease-related genes through analysis of gene network topology

    • Apply algorithms that find more disease-related genes than comparable methods

  • Text Mining Strategies:

    • Utilize co-occurrence-based approaches to identify relationships between LIF and other biological entities

    • Implement opinion sentence analysis to infer describable disease-gene relationships

    • Apply specialized strategies like TILD (Title Information in Literature Data) to identify cancer-related genes

  • Data Integration:

    • Combine expression data with protein-protein interaction networks

    • Incorporate pathway enrichment analysis

    • Utilize machine learning algorithms to predict functional relationships

These methodologies collectively enable researchers to systematically analyze complex relationships between LIF expression and disease phenotypes, facilitating hypothesis generation for experimental validation.

What role does LIF play in stem cell pluripotency networks and cellular reprogramming?

LIF functions as a critical regulator within pluripotency networks, with significant implications for cellular reprogramming:

  • Signaling Pathway Integration: LIF signaling interfaces with other pluripotency pathways, particularly through STAT3 activation, which cooperates with core pluripotency factors OCT4, SOX2, and NANOG.

  • Epigenetic Remodeling: LIF induces specific epigenetic modifications, including changes in DNA methylation and histone modifications at pluripotency-associated gene loci.

  • Convergence with bFGF Signaling: Recent evidence suggests that LIF and bFGF signaling pathways, despite their distinct receptor activation mechanisms, converge on common OCT4 target genes, challenging previous assumptions about cytokine-defined stem cell phenotypes .

  • Enhanced Reprogramming Efficiency: When combined with transcriptionally enhanced OCT4 (M³O), LIF contributes to significantly improved reprogramming efficiency and reduced time requirements for iPSC generation .

  • Naïve-to-Primed State Transitions: LIF plays a key role in maintaining cells in a more naïve pluripotent state, though the distinctions between naïve and primed states appear more complex than previously thought.

This research area continues to evolve, with ongoing investigations into the precise mechanisms by which LIF influences the pluripotency network and facilitates cellular reprogramming.

How can single-cell analysis enhance our understanding of heterogeneous LIF responses in mixed cell populations?

Single-cell technologies offer powerful approaches to dissect heterogeneous responses to LIF stimulation:

  • Single-Cell RNA Sequencing (scRNA-seq): Enables identification of cell subpopulations with distinct LIF response signatures and reveals trajectory-dependent responses during differentiation or reprogramming.

  • Single-Cell Protein Analysis: Techniques such as mass cytometry (CyTOF) and single-cell western blotting provide protein-level insights into LIF signaling at individual cell resolution.

  • Spatial Transcriptomics: Methods like MERFISH or Visium spatial gene expression profiling maintain spatial context while revealing LIF response patterns across tissue architecture.

  • Live Cell Imaging: Fluorescent reporters for LIF pathway activity enable dynamic tracking of signaling responses in individual cells over time.

  • Computational Integration: Advanced algorithms combining data across platforms can reconstruct LIF-dependent gene regulatory networks at single-cell resolution.

These approaches collectively overcome limitations of bulk analysis methods, which may obscure important cell-to-cell variations in LIF responsiveness that influence cellular fate decisions and function.

What are the most common pitfalls in LIF-based experiments and how can researchers address them?

Researchers frequently encounter several challenges when working with LIF in experimental systems:

ChallengeCauseSolution
Inconsistent stem cell maintenanceVariability in LIF activityUse standardized bioactivity assays; maintain consistent lot numbers; consider recombinant LIF with defined activity units
Non-specific antibody binding in LIF detectionCross-reactivity with other IL-6 family cytokinesPerform validation with positive and negative controls; use multiple antibodies targeting different epitopes
Contradictory results across cell linesCell type-specific LIF receptor expressionQuantify LIFR and gp130 levels before experiments; normalize data to receptor expression
Failure to replicate published findingsDifferences in experimental conditionsCarefully match media composition, passage number, cell density, and timing of analysis
Batch effects in RNA-seq analysisTechnical variationInclude batch correction in analysis pipeline; use spike-in controls; perform experiments in parallel

Additional methodological considerations include:

  • For LIF-dependent stem cell culture, regular monitoring of pluripotency markers is essential

  • When comparing LIF to other cytokines, ensure equivalent bioactivity rather than relying solely on concentration

  • In therapeutic antibody development against LIF, confirm target engagement using both in vitro and in vivo validation methods

Product Science Overview

Discovery and History

LIF was discovered in the late 1980s by researchers at the Walter and Eliza Hall Institute in Melbourne, Australia. They isolated a protein from mouse Krebs tumor cells that could induce the differentiation of mouse myeloid leukemia cells into macrophages and granulocytes without promoting their proliferation . This protein was named Leukemia Inhibitory Factor due to its inhibitory effects on leukemia cell proliferation .

Structure and Expression

LIF is a glycoprotein with a molecular weight of approximately 20 kDa . It is expressed in various tissues and cell types, including the trophectoderm of the developing embryo, where it plays a crucial role in implantation and early embryonic development . The LIF receptor (LIFR) is a heterodimer composed of LIFR (gp190) and gp130, a common signal transducer for IL-6-type cytokines .

Biological Functions

LIF has a wide range of biological functions, including:

  • Inducing differentiation: LIF can induce the terminal differentiation of various cell types, including myeloid leukemia cells and embryonic stem cells .
  • Maintaining stem cell pluripotency: LIF is commonly used in the laboratory to maintain the undifferentiated state of mouse embryonic stem cells .
  • Regulating immune responses: LIF can modulate immune responses by influencing the activity of various immune cells .
  • Promoting cell survival: LIF has been shown to promote the survival of neurons and other cell types under stress conditions .
Recombinant Human LIF

Recombinant human LIF (hLIF) is produced using recombinant DNA technology, typically in bacterial or mammalian cell expression systems . It is used extensively in research and biotechnology for its ability to maintain the pluripotency of embryonic stem cells and its various other biological activities .

Applications in Research and Medicine
  • Stem cell research: hLIF is widely used to maintain the undifferentiated state of mouse embryonic stem cells in culture .
  • Cancer research: Due to its role in cell differentiation and survival, hLIF is studied for its potential therapeutic applications in cancer treatment .
  • Neurobiology: hLIF is investigated for its neuroprotective effects and potential use in treating neurodegenerative diseases .

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.