LIF Human, Yeast

LIF Human Recombinant, Yeast
Shipped with Ice Packs
In Stock

Description

Production and Quality Control

The protein is synthesized using proprietary chromatographic techniques in Pichia pastoris, followed by lyophilization. Critical quality metrics include:

  • Biological Activity: Tested via murine M1 myeloid leukemic cell differentiation assays, showing a specific activity of >1 × 10⁸ units/mg .

  • Endotoxin Levels: <1.0 EU/µg, ensuring suitability for cell culture .

Mechanism of Action

LIF Human, Yeast binds to the LIF receptor (LIFR) and gp130, activating downstream pathways such as JAK-STAT (primarily STAT3) and MAPK/ERK. This signaling:

  • Maintains embryonic stem cell pluripotency by suppressing differentiation .

  • Modulates immune responses and tumor progression (e.g., pancreatic cancer) by interacting with stromal myofibroblasts .

Stem Cell Research

  • Pluripotency Maintenance: LIF suppresses spontaneous differentiation in embryonic stem cells, enabling long-term culture .

  • Therapeutic Potential: Supports regenerative medicine for tissue repair and infertility treatments (e.g., improving IVF implantation rates) .

Cancer Studies

  • Ligand Trap Development: Engineered LIFR-Fc fusion proteins (e.g., eLIFR-Fc) inhibit LIF signaling, reducing tumor growth in pancreatic cancer models .

  • Targeted Therapy: Blocks KRAS-driven pathways in pancreatic ductal adenocarcinoma (PDAC) .

Comparative Analysis with Other LIF Variants

FeatureLIF Human, YeastE. coli-Derived LIFMammalian Cell-Derived LIF
GlycosylationYesNoYes
Molecular Mass58.5 kDa~20 kDa~38–58 kDa
CostModerateLowHigh
ApplicationsStem cell/cancer studiesBasic researchTherapeutic development
Source

Emerging Therapeutic Strategies

Recent studies highlight engineered LIF inhibitors (e.g., eLIFR-Fc) with ~20 pM affinity for LIF, showing efficacy in preclinical cancer models without significant toxicity . These traps compete with native receptors, blocking protumorigenic signaling .

Product Specs

Introduction
Leukemia Inhibitory Factor (LIF) is a lymphoid factor that plays a crucial role in maintaining embryonic stem cells in their undifferentiated state by preventing spontaneous differentiation. LIF exhibits various functions, including promoting the differentiation of cholinergic neurons, regulating stem cell pluripotency, influencing bone and fat metabolism, stimulating the growth of factor-dependent cell lines, and enhancing the production of megakaryocytes in living organisms. Notably, there is a high degree of similarity between human and mouse LIF, with 78% identical amino acid sequences.
Description
Recombinant Human LIF, produced in yeast, is a single-chain polypeptide that has undergone glycosylation. It comprises 180 amino acids and has a molecular weight of 58.5 kDa. The purification process for LIF involves specialized chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder that has been sterilized by filtration.
Formulation
The protein was lyophilized from a 0.2 μm filtered phosphate-buffered saline (PBS) solution.
Solubility
To reconstitute the lyophilized LIF, it is advised to dissolve it in sterile, 18 megaohm-centimeter (MΩ·cm) H₂O at a concentration of at least 100 μg/mL. This solution can then be further diluted into other aqueous solutions as needed.
Stability
Lyophilized LIF remains stable for up to 3 weeks at room temperature. However, for long-term storage, it is recommended to store the desiccated product at temperatures below -18°C. Once reconstituted, LIF can be stored at 4°C for 2-7 days. For longer storage periods, it should be kept at temperatures below -18°C. It is crucial to avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of the protein is greater than 98.0%, as determined by the following methods: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis, and (b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity of recombinant human LIF was evaluated based on its ability to induce the differentiation of murine M1 myeloid leukemia cells. The minimum concentration of human LIF required to elicit a detectable response in this assay is less than 0.05 ng/mL. The specific activity of the protein is greater than 1 x 10⁸ units/mg.
Synonyms
CDF, HILDA, D-FACTOR, Differentiation- stimulating factor, Melanoma-derived LPL inhibitor, MLPLI, Emfilermin, Leukemia inhibitory factor, LIF, DIA.
Source
Pichia pastoris.
Amino Acid Sequence
S P L P I T P V N A T C A I R H P C H N N L M N Q I R S Q L A Q L N G S A N A L F I L Y Y T A Q G E P F P N N L D K L C G P N V T D F P P F H A N G T E K A K L V E L Y R I V V Y L G T S L G N I T R D Q K I L N P S A L S L H S K L N A T A D I L R G L L S N V L C R L C S K Y H V G H V D V T Y G P D T S G K D V F Q K K K L G C Q L L G K Y K Q I I A V L A Q A F.

Q&A

What is the evolutionary relationship between human and yeast genes?

Despite approximately one billion years of evolutionary divergence, humans and baker's yeast (Saccharomyces cerevisiae) share hundreds of conserved genes that remain functionally similar. Research has demonstrated that these genes originate from a common ancestor and have maintained their critical cellular functions despite significant species divergence . This conservation is particularly evident in fundamental cellular processes such as metabolism, DNA replication, and protein synthesis. Notably, of the approximately 450 genes critical for yeast survival that have human counterparts, nearly half can be functionally replaced with their human versions while maintaining yeast viability . This remarkable conservation provides powerful evidence for the common heritage of all living organisms and creates valuable opportunities for using yeast as model systems for human gene function studies.

What defines "humanized yeast" and what are its primary research applications?

Humanized yeast refers to genetically engineered yeast strains where native yeast genes have been systematically replaced with their human counterparts. These engineered organisms are created by removing specific yeast genes and introducing the corresponding human genes, then assessing whether the humanized yeast can survive and reproduce normally . The primary research applications include:

  • Studying fundamental gene function conservation between species

  • Investigating the effects of human gene mutations in a simplified cellular system

  • Screening potential therapeutic compounds for genetic disorders

  • Exploring evolutionary biology questions regarding gene conservation

  • Developing platforms for drug discovery and validation

Researchers at the University of Texas at Austin have demonstrated that nearly half of the critical yeast genes tested could be successfully replaced with human versions, creating viable humanized yeast strains . These engineered organisms provide unique opportunities to study human gene function in a controlled, single-celled system that is easier to manipulate and analyze than human cells or animal models.

How do researchers determine if a human gene can functionally replace its yeast counterpart?

Determining functional replacement involves a systematic experimental approach:

  • Gene identification and isolation: Researchers first identify corresponding gene pairs between yeast and humans based on sequence similarity and predicted function .

  • Targeted gene deletion: The native yeast gene is precisely removed from the yeast genome using molecular techniques such as homologous recombination .

  • Human gene introduction: The human gene is cloned into a yeast expression vector and introduced into the modified yeast strain .

  • Viability assessment: Researchers observe whether the engineered yeast strain can survive and reproduce normally with only the human version of the gene .

  • Functional verification: Additional tests assess whether specific cellular functions are maintained, including growth rate measurement, response to environmental stresses, and biochemical pathway functionality .

  • Comparative analysis: Data is analyzed to determine which genes are successfully complemented and which fail, providing insights into functional conservation versus divergence .

This methodology has been successfully applied to hundreds of gene pairs, with approximately 47% of critical yeast genes being successfully replaced by their human counterparts in studies conducted at the University of Texas at Austin .

What techniques are used to engineer high-affinity LIF receptors (LIFRs) for research applications?

Engineering high-affinity LIFR variants involves sophisticated protein engineering techniques:

  • Domain identification: Researchers first identify the minimal binding domains required for LIF interaction. For human LIFR, the three N-terminal domains (CBM I, Ig-like, and CBM II) contribute to LIF binding, with the Ig-like domain mediating most contacts .

  • Yeast surface display: The extracellular domains of LIFR are expressed on yeast cell surfaces, creating a platform for directed evolution experiments .

  • Error-prone PCR mutagenesis: Targeted mutagenesis introduces random mutations, particularly in the Ig-like domain, to generate diverse LIFR variant libraries (approximately 2.7 × 10^7 transformants in initial studies) .

  • Fluorescence-activated cell sorting (FACS): Multiple rounds of increasingly stringent sorting isolate yeast displaying LIFR variants with enhanced binding to LIF .

  • DNA shuffling: Beneficial mutations from first-generation variants are recombined to create second-generation libraries with improved binding properties .

  • Affinity measurement: Techniques like Kinetic Exclusion Assay (KinExA) quantify binding affinity improvements between engineered variants and wild-type receptors .

This approach has yielded engineered LIFR variants with approximately 50-fold increased affinity (~20 pM) compared to wild-type LIFR, demonstrating the power of directed evolution for protein engineering .

How can humanized yeast models be optimized for studying human genetic disorders?

Optimizing humanized yeast for genetic disorder studies requires several methodological considerations:

  • Selection of appropriate gene pairs: Focus on disease-relevant genes with clear yeast orthologs that can be functionally replaced by human versions .

  • Introduction of disease-specific mutations: Engineer precise mutations in human genes that mirror those found in patients with genetic disorders .

  • Phenotypic readout development: Establish assays that can detect functional consequences of mutations, such as growth defects, metabolic alterations, or protein mislocalization .

  • Strain engineering for human-like cellular context: Modify yeast cellular environment when necessary to better mimic human cellular conditions (e.g., introducing human chaperones, modifying post-translational modification machinery) .

  • High-throughput screening platforms: Develop miniaturized assay formats compatible with automated screening to test multiple mutations or compound libraries .

  • Validation in mammalian systems: Confirm findings from yeast models in human cell lines or animal models to establish clinical relevance .

  • Computational integration: Combine experimental data with bioinformatic approaches to predict mutation effects and identify potential therapeutic targets .

This systematic approach enables researchers to create yeast-based platforms for studying specific genetic disorders and screening for compounds that might restore normal function, as demonstrated in studies of various human diseases .

What methods are employed to assess the binding affinity and inhibitory potential of engineered LIF receptor variants?

Researchers employ multiple complementary techniques to rigorously characterize engineered LIFR variants:

  • Flow cytometry binding assays: Quantify binding of fluorescently labeled LIF to LIFR variants displayed on yeast cell surfaces under various concentration conditions to determine apparent dissociation constants (Kd) .

  • Competition binding assays: Measure displacement of bound LIF by soluble competitors to assess relative binding strengths and off-rates of different LIFR variants .

  • Kinetic Exclusion Assay (KinExA): Determine precise binding affinities between soluble LIFR fusion proteins and LIF using solution-phase interactions without surface artifacts .

  • Phospho-STAT3 inhibition assays: Measure the ability of LIFR variants to prevent LIF-induced STAT3 phosphorylation in cancer cell lines, providing a functional readout of inhibitory potential .

  • Fc-fusion protein engineering: Create bivalent LIFR-Fc fusion proteins to enhance avidity effects and extend serum half-life for improved inhibitory properties .

  • Sphere formation assays: Assess inhibition of LIF-dependent cellular phenotypes, such as cancer cell sphere formation, to validate biological relevance .

  • In vivo tumor models: Evaluate the effect of engineered LIFR variants on tumor progression in animal models to confirm therapeutic potential .

These complementary approaches have demonstrated that engineered LIFR variants with ~50-fold improved binding affinity show enhanced inhibition of LIF signaling and promising therapeutic potential .

What patterns of gene conservation have been observed between humans and yeast?

Research on gene conservation between humans and yeast has revealed several significant patterns:

  • Functional category bias: Genes involved in core cellular processes (metabolism, protein synthesis, DNA replication) show higher conservation and replaceability than those involved in species-specific functions .

  • Structural conservation despite sequence divergence: Many genes maintain functional compatibility despite considerable amino acid sequence differences, suggesting structural and mechanistic conservation is more important than sequence identity .

  • Module-based conservation: Entire functional modules or protein complexes tend to be either highly conserved or highly divergent as complete units, rather than showing mixed conservation patterns .

  • Evolutionary rate consistency: Genes that evolve slowly in one lineage tend to evolve slowly in the other lineage as well, indicating consistent selection pressures on critical functions .

  • Replaceability correlations: Nearly half (approximately 47%) of the essential yeast genes tested could be functionally replaced by their human counterparts, suggesting remarkable functional conservation across a billion years of evolution .

These patterns provide profound insights into the evolutionary constraints on gene function and the core machinery of eukaryotic cells that has remained largely unchanged since the last common ancestor of humans and yeast .

How do researchers identify and validate gene pairs between humans and yeast for functional replacement studies?

The identification and validation of orthologous gene pairs involves a multi-step process:

  • Sequence-based identification: Initial ortholog pairs are identified using sequence similarity algorithms (BLAST, HMMER) and existing ortholog databases (OrthoMCL, InParanoid) .

  • Structural homology assessment: Protein structure prediction and comparison helps identify functional equivalence even when sequence similarity is low .

  • Domain architecture analysis: Comparison of protein domain organization helps confirm orthology relationships and identify core functional regions .

  • Phylogenetic reconstruction: Detailed evolutionary history analysis distinguishes true orthologs from paralogs resulting from gene duplications .

  • Literature-based functional annotation: Manual curation of known gene functions helps identify the most promising candidates for replacement studies .

  • Experimental validation: The ultimate test is the functional replacement experiment itself, where the yeast gene is deleted and the human gene is introduced to test for viability .

  • Large-scale screening: Systematic testing of hundreds of gene pairs provides statistical power to identify patterns of replaceability and conservation .

This methodical approach has enabled researchers to systematically analyze approximately 450 essential yeast genes and their human counterparts, revealing that around 200 human genes can successfully replace their yeast equivalents .

How can humanized yeast facilitate drug discovery for human genetic disorders?

Humanized yeast provides several unique advantages for drug discovery applications:

  • Simplified screening system: Yeast provides a eukaryotic cellular environment without the complexity of multicellular organisms, enabling high-throughput drug screening .

  • Disease mutation modeling: Researchers can introduce precise versions of human gene mutations into yeast and test multiple drug candidates to identify potential treatments .

  • Personalized medicine applications: Different versions of a human gene mutation can be tested in parallel to determine which treatments might work best for specific genetic variants .

  • Target validation: Before investing in mammalian studies, researchers can verify that compounds directly affect the function of specific disease-associated proteins .

  • Mechanism of action studies: The simplified yeast system facilitates understanding how compounds interact with target proteins and affect cellular pathways .

  • Combination therapy screening: Multiple compounds can be tested simultaneously to identify synergistic effects that might not be apparent in more complex systems .

  • Off-target effect reduction: Initial screening in yeast can help identify compounds with specific effects on the target of interest before moving to human cell testing .

As noted by researchers at the University of Texas at Austin, these advantages make humanized yeast valuable for searching for drugs to treat genetic diseases, potentially identifying approximately 1,000 additional gene pairs that might be useful for drug screening applications .

What are the mechanisms and applications of engineered LIFR decoys as therapeutic agents?

Engineered LIFR decoys represent a promising therapeutic strategy through several mechanisms:

  • Ligand sequestration mechanism: Engineered LIFR variants act as "ligand traps" by binding to and sequestering LIF, preventing it from activating endogenous receptors on cell surfaces .

  • Affinity enhancement: Through directed evolution and protein engineering, LIFR variants with dramatically increased binding affinity (~20 pM, 50-fold higher than wild-type) more effectively compete with native receptors for LIF binding .

  • Signal pathway inhibition: By preventing LIF-receptor interaction, these decoys block downstream signaling pathways, including STAT3 phosphorylation, which is implicated in cancer progression .

  • Fc-fusion design: Fusion to antibody Fc domains creates bivalent constructs with enhanced avidity, extended serum half-life, and improved pharmacokinetic properties .

  • Cancer therapy applications: LIFR decoys show promise in inhibiting LIF-mediated effects in pancreatic cancer cells and slowing xenograft tumor growth .

  • Reduced immunogenicity: As modified versions of human proteins, these decoys potentially offer lower immunogenicity compared to antibody-based therapeutics .

  • Platform extensibility: The directed evolution approach used for LIFR engineering could be applied to other cytokine receptors for developing similar therapeutic strategies .

Experimental validation has demonstrated that engineered LIFR-Fc constructs effectively block LIF-derived signaling and sphere formation in pancreatic ductal adenocarcinoma (PDAC) cells and slow tumor progression in mouse models, highlighting their therapeutic potential .

What limitations must researchers address when using humanized yeast models?

Despite their utility, humanized yeast models present several technical challenges:

  • Cellular context differences: Yeast lacks many human-specific cellular components, potentially affecting the function of some human proteins .

  • Post-translational modification variations: Differences in protein modification systems between yeast and humans may impact protein function and interactions .

  • Metabolic pathway divergence: Certain metabolic pathways have diverged significantly, limiting the utility of yeast for studying human-specific metabolism .

  • Absence of multicellularity: Yeast cannot model tissue-specific or developmental processes requiring cell-cell interactions .

  • Non-replaceable genes: Approximately 53% of essential yeast genes cannot be functionally replaced by their human counterparts, indicating significant functional divergence in these cases .

  • Limited modeling of membrane and extracellular proteins: Differences in membrane composition and extracellular environment may affect the function of these protein classes .

  • Gene dosage sensitivity: Some human genes may require precise expression levels that are difficult to achieve in yeast expression systems .

Researchers must carefully consider these limitations when designing experiments and interpreting results from humanized yeast models, often necessitating validation in mammalian systems for translational relevance .

What emerging technologies are advancing the field of human-yeast comparative genomics?

Several cutting-edge technologies are accelerating progress in human-yeast comparative genomics:

  • CRISPR-Cas9 genome editing: Enables precise and efficient modification of both yeast and human genomes, facilitating systematic humanization studies .

  • Single-cell analysis techniques: Allow researchers to assess the variability in humanized yeast populations and identify subtle phenotypic effects .

  • High-throughput phenotyping: Automated growth measurement, microscopy, and metabolic profiling enable comprehensive characterization of humanized yeast strains .

  • Protein structure prediction advances: Tools like AlphaFold2 improve the identification of functionally equivalent proteins despite low sequence similarity .

  • Synthetic genomics approaches: Working toward creating yeast strains with multiple chromosomes replaced by human counterparts for more comprehensive modeling .

  • Multi-omics integration: Combining transcriptomics, proteomics, and metabolomics data provides holistic understanding of humanized yeast function .

  • Computational modeling and simulation: Predicting the effects of human gene substitutions and guiding experimental design through in silico approaches .

These technological advances are expanding the scope and resolution of human-yeast comparative studies, potentially leading to the identification of approximately 1,000 additional swappable gene pairs beyond those already characterized .

Product Science Overview

Production and Characteristics

Human Recombinant LIF produced in yeast is a single, glycosylated polypeptide chain containing 180 amino acids and has a molecular mass of approximately 58.5 kDa . The expression host for this recombinant protein is typically Pichia pastoris, a species of yeast known for its ability to perform post-translational modifications similar to those in higher eukaryotes .

The recombinant LIF is purified using proprietary chromatographic techniques to ensure high purity, typically greater than 98% as determined by RP-HPLC and SDS-PAGE . The protein is lyophilized from a 0.2 µm filtered PBS solution and is recommended to be reconstituted in sterile water to a concentration of at least 100 µg/ml .

Biological Functions

LIF has several important biological functions, including:

  • Maintenance of Embryonic Stem Cells: LIF is crucial for the long-term maintenance of embryonic stem cells by suppressing spontaneous differentiation .
  • Neuronal Differentiation: It promotes the differentiation of cholinergic neurons .
  • Bone and Fat Metabolism: LIF plays a role in the regulation of bone and fat metabolism .
  • Hematopoiesis: It is involved in the mitogenesis of factor-dependent cell lines and the promotion of megakaryocyte production in vivo .
  • Inflammation: LIF has a role in inflammatory responses .
Applications

Recombinant human LIF is widely used in research and biotechnology for various applications, including:

  • Stem Cell Research: LIF is used to maintain the pluripotency of embryonic stem cells and induced pluripotent stem cells (iPSCs) in culture .
  • Differentiation Studies: It is employed in studies investigating the differentiation of various cell types .
  • Functional Assays: LIF is used in functional assays to study its effects on different biological processes .
Stability and Storage

Lyophilized LIF is stable at room temperature for up to three weeks but should be stored desiccated below -18°C for long-term storage . Upon reconstitution, it should be stored at 4°C for short-term use (2-7 days) and below -18°C for future use. It is important to avoid freeze-thaw cycles to maintain the protein’s stability .

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.