MIF Human His C is a non-glycosylated polypeptide with a His-tag appended to its C-terminus. This modification facilitates affinity chromatography purification using nickel or cobalt resin.
The His-tag does not interfere with MIF’s native trimeric structure or enzymatic activity. Structural studies confirm that the C-terminal His-tag is positioned away from the active sites (N-terminal phenylpyruvate tautomerase motif and C-terminal thiol-protein oxidoreductase motif) .
MIF Human His C is produced via recombinant expression in E. coli. Key steps include:
Cloning: Insertion of the MIF gene into a plasmid with a C-terminal His-tag coding sequence.
Expression: Induced in E. coli under optimized conditions.
Purification: Affinity chromatography (Ni-NTA or Co columns) followed by size-exclusion chromatography (SEC) to ensure homogeneity .
Purity Assessment | Method | Result | Source |
---|---|---|---|
SDS-PAGE | Single band at ~13.5 kDa | >95% purity | |
HPLC | RP-HPLC analysis | >95% purity | |
Endotoxin Level | LAL assay | <1 EU/μg |
MIF Human His C retains the biological activities of native MIF, including:
Cytokine Activity: Proinflammatory signaling via CD74 and CD44 receptors, promoting NF-κB activation and cytokine (e.g., TNF-α, IL-8) production .
Enzymatic Activity:
Glucocorticoid Antagonism: Counteracts immunosuppressive effects of steroids by stabilizing IκBα and preventing NF-κB inhibition .
MIF Human His C binds to CD74 with high specificity, as validated in functional ELISAs. This interaction is critical for its role in immune modulation and tumor progression .
A CATT repeat polymorphism in the MIF promoter (-794 CATT) influences MIF expression levels. High-repeat alleles are associated with increased disease severity in rheumatoid arthritis and tuberculosis .
Small-molecule inhibitors (e.g., coumarin derivatives) targeting MIF’s tautomerase active site show IC₅₀ values <5 µM, highlighting potential for anti-inflammatory therapies .
MIF (Macrophage Migration Inhibitory Factor) is a pro-inflammatory cytokine that functions as an integral component of the host antimicrobial alarm system and stress response. Originally discovered in the late 1960s as a product of activated T cells, MIF has since been recognized as a crucial mediator of innate immunity . It is constitutively expressed by a broad spectrum of cells and tissues, including monocytes and macrophages, and is rapidly released following exposure to microbial products, pro-inflammatory mediators, and in response to stress .
The primary biological functions of MIF include:
Activation of the ERK1/ERK2-mitogen-activated protein kinase pathway
Inhibition of JUN activation domain-binding protein 1 (JAB1)
Upregulation of Toll-like receptor 4 expression to enhance recognition of endotoxin-expressing bacterial pathogens
Sustaining pro-inflammatory functions by inhibiting p53-dependent apoptosis of macrophages
Counter-regulating the immunosuppressive effects of glucocorticoids on immune cells
Expression and purification of MIF typically follows these methodological steps:
Gene cloning: The MIF coding sequence is amplified using PCR and cloned into an expression vector containing a histidine tag. The MIF promoter region (approximately 1kb fragment) can be derived from GenBank AF033192, as demonstrated in some studies .
Expression system selection: Most commonly, recombinant human MIF (rhMIF) is expressed in bacterial systems such as E. coli, though mammalian expression systems may be used for studies requiring post-translational modifications.
Protein induction: Expression is induced using IPTG or other appropriate inducers depending on the expression system chosen.
Cell lysis: Bacterial cells are lysed using mechanical disruption or chemical methods to release the recombinant protein.
Purification: His-tagged MIF protein is typically purified using nickel or cobalt affinity chromatography, exploiting the affinity of the histidine tag for metal ions.
Quality control: The purified protein should undergo verification by SDS-PAGE, Western blotting, and activity assays to confirm identity, purity, and biological activity.
Several reliable methods exist for detecting and quantifying MIF in biological samples, each with specific advantages:
Enzyme-Linked Immunosorbent Assay (ELISA): This is the most common method for quantifying MIF in serum, plasma, or cell culture supernatants. Standard ELISA protocols utilize a coating antibody (e.g., monoclonal anti-MIF MAB289) and a biotinylated anti-human MIF capture antibody (e.g., goat anti-human, BAF289) . This method offers high sensitivity and specificity for MIF quantification.
Western Blotting: This technique is suitable for detecting MIF in cell or tissue lysates, allowing for protein size confirmation. Studies have used this method to verify MIF expression in various cell types, including hepatic sinusoidal endothelial cells .
Immunohistochemistry: For tissue samples, immunohistochemistry using anti-MIF polyclonal antibodies (such as those from R&D Systems) followed by biotinylated secondary antibodies and ABC-peroxidase reagent with appropriate substrate can visualize MIF expression patterns in situ .
Real-time quantitative PCR (qPCR): For measuring MIF gene expression at the mRNA level, qPCR has been effectively used to verify MIF expression in various experimental models .
Cytokine Arrays: For multiplex analysis, human cytokine arrays containing antibodies against MIF alongside other cytokines can provide semi-quantitative comparison of MIF levels across different samples .
MIF plays complex roles in hepatocellular carcinoma (HCC) pathogenesis through multiple mechanisms:
Genetic susceptibility: MIF promoter polymorphisms, particularly rs755622, are associated with increased susceptibility to HCC. The GC and CC genotypes at this locus correlate with elevated MIF protein expression in peripheral blood and poorer prognosis compared to the GG genotype .
Diagnostic biomarker potential: Studies have demonstrated that MIF expression is significantly increased in the peripheral blood of HCC patients compared to healthy individuals, chronic hepatitis B patients, and liver cirrhosis patients .
Role in metastasis: In colorectal cancer liver metastasis, MIF secreted by human hepatic sinusoidal endothelial cells (HHSECs) promotes:
Molecular mechanisms: MIF accelerates mobility of cancer cells by suppressing F-actin depolymerization and phosphorylating cofilin. Additionally, MIF correlates with the size of hepatic metastases, suggesting its role in tumor growth .
In vivo evidence: Orthotopic implantation models in nude mice have shown that exogenous MIF stimulates growth of colorectal cancer cells and metastasis. Tumors arising from implantation of cancer cells mixed with MIF-secreting cells showed higher growth rates, volumes, weights, and metastatic foci compared to controls .
Distinguishing between autocrine and paracrine effects of MIF requires careful experimental design:
Cell-specific knockdown strategies:
Utilizing shRNA to selectively knock down MIF in specific cell types can help determine source-dependent effects. For example, studies have used lentiviral vector-mediated shRNA expression to inhibit MIF in hepatic sinusoidal endothelial cells (HHSECs) while maintaining MIF expression in cancer cells .
Complementary experiments involving MIF knockdown in cancer cells while maintaining MIF expression in stromal cells can further delineate the source-dependent effects.
Conditioned media experiments:
Collecting conditioned media from different cell types (with or without MIF knockdown) and applying it to recipient cells can help identify paracrine effects.
For example, researchers have cultured colorectal cancer cells with conditioned media from HHSECs with varying MIF expression levels to demonstrate that MIF released from HHSECs, rather than from cancer cells themselves, promotes migration, EMT, proliferation, and apoptotic resistance .
Co-culture systems with selective inhibition:
Using Transwell systems to physically separate different cell types while allowing soluble factor exchange
Applying MIF inhibitors (such as ISO-1 or P425) selectively to either compartment can help determine directional effects
Studies have shown that application of MIF inhibitors resulted in inhibition of HHSEC-induced migration of colorectal cancer cells
Recombinant protein rescue experiments:
MIF polymorphisms significantly impact protein expression and disease susceptibility through several mechanisms:
Promoter region variations: The MIF rs755622 polymorphism (-173 G/C) in the promoter region has been associated with altered MIF expression levels. Specifically:
Disease progression correlation: MIF promoter polymorphisms not only affect disease susceptibility but also influence disease progression and outcomes:
Quantitative differences: Studies measuring MIF protein levels in peripheral blood have found:
Experimental verification: The relationship between MIF promoter polymorphisms and expression can be verified experimentally using:
Optimal conditions for expressing and purifying biologically active MIF-His tagged protein include:
Expression vector selection:
pET expression systems (e.g., pET21a) are commonly used for high-level expression
The His-tag position (N- or C-terminal) should be carefully chosen; C-terminal tags often minimize interference with the MIF active site
Expression conditions:
Host strain: BL21(DE3) E. coli is typically preferred for high yield
Induction: 0.5-1.0 mM IPTG at OD600 0.6-0.8
Temperature: Lower temperatures (16-25°C) after induction often improve solubility
Duration: 4-16 hours of induction depending on temperature
Cell lysis:
Buffer composition: 50 mM Tris-HCl pH 8.0, 150 mM NaCl, 1 mM EDTA, protease inhibitors
Method: Sonication or high-pressure homogenization for efficient lysis
Centrifugation: High-speed centrifugation (≥20,000 × g) to remove cellular debris
Purification strategy:
Primary purification: Ni-NTA or TALON (cobalt-based) affinity chromatography
Buffer conditions: 50 mM sodium phosphate pH 8.0, 300 mM NaCl, 10 mM imidazole for binding
Elution: Step gradient of imidazole (50-250 mM)
Secondary purification: Size exclusion chromatography to remove aggregates and ensure homogeneity
Quality control:
SDS-PAGE to assess purity
Western blot using anti-His and anti-MIF antibodies
Endotoxin removal: Critical for immunological assays, using polymyxin B columns or commercial endotoxin removal kits
Activity testing: Tautomerase activity assay using L-dopachrome methyl ester as substrate
Storage conditions:
Buffer: PBS or 20 mM Tris-HCl pH 7.4, 150 mM NaCl
Addition of 5-10% glycerol increases stability
Storage at -80°C in small aliquots to avoid freeze-thaw cycles
Researchers can measure MIF functional activity using various methodological approaches tailored to different experimental settings:
Enzymatic activity assays:
Tautomerase activity: MIF exhibits tautomerase activity that can be measured using L-dopachrome methyl ester as a substrate
Phenylpyruvate tautomerase activity: Using p-hydroxyphenylpyruvate as substrate
These assays provide quick verification of proper protein folding and enzymatic function
Cell migration assays:
Transwell migration assays: These have been extensively used to assess MIF's chemotactic effects on various cell types including colorectal cancer cells
Wound healing (scratch) assays: To measure the role of MIF in cell migration and wound closure
Inhibition studies using specific MIF inhibitors (ISO-1, P425) can confirm specificity
Signal transduction analysis:
Gene expression modulation:
In vivo functional assays:
Receptor binding and activation:
CD74, CXCR2, and CXCR4 are known MIF receptors
Flow cytometry or surface plasmon resonance to measure binding
Receptor signaling assays to assess downstream activation
Analyzing contradictory data about MIF functions across different disease contexts presents several methodological challenges:
Context-dependent effects:
MIF exhibits distinct roles in different cellular contexts and disease states
For example, while MIF promotes tumor growth in many cancers , it may have protective effects in certain inflammatory conditions
Methodological approach: Use matched cell types and tissue samples when comparing across studies; explicitly state the disease context and experimental conditions in publications
Source-dependent variations:
MIF derived from different cellular sources may have distinct effects
Studies have shown that MIF released by hepatic sinusoidal endothelial cells, but not by colorectal cancer cells themselves, promotes migration and EMT
Methodological approach: Clearly identify and characterize the source of MIF in experiments; use cell-specific knockdown or knockout models
Concentration-dependent effects:
MIF may exhibit hormetic responses where low and high concentrations produce opposite effects
Methodological approach: Use concentration gradients in experiments; report precise concentrations used; standardize recombinant protein preparations
Receptor expression heterogeneity:
Different cell types express varying levels of MIF receptors (CD74, CXCR2, CXCR4)
Methodological approach: Characterize receptor expression in experimental models; consider receptor blocking experiments to identify which receptor mediates specific effects
Post-translational modifications:
MIF function may be altered by oxidation, glycation, or other modifications
Methodological approach: Characterize post-translational modifications of MIF in different experimental systems; use mass spectrometry to identify modifications
Genetic variation:
MIF polymorphisms affect expression levels and possibly protein function
The MIF rs755622 polymorphism influences susceptibility to hepatocellular carcinoma and correlates with protein expression levels
Methodological approach: Genotype subjects or cell lines for known MIF polymorphisms; stratify analyses based on genotype
Temporal dynamics:
MIF may have different effects during acute versus chronic phases of disease
Methodological approach: Design time-course experiments; compare acute and chronic models of the same disease
MIF plays multiple crucial roles in shaping the tumor microenvironment during liver metastasis through several mechanisms:
Promotion of cancer cell chemotaxis:
Induction of epithelial-mesenchymal transition (EMT):
Cancer cells exposed to MIF-rich conditioned media from HHSECs undergo morphological changes, appearing "starfish shaped" with cytoplasmic protuberances
Molecularly, MIF stimulates increased expression of mesenchymal markers (N-cadherin, vimentin) and decreased expression of epithelial markers (E-cadherin)
This EMT phenotype enhances cancer cell invasiveness and mobility within the liver microenvironment
Cytoskeletal regulation:
Promotion of cancer cell proliferation and survival:
In the liver microenvironment, MIF enhances cancer cell proliferation as demonstrated by EdU assays and CCK8 proliferation assays
MIF also confers apoptotic resistance to cancer cells, promoting their survival in the new microenvironment
These effects are predominantly mediated by MIF secreted from HHSECs rather than autocrine MIF from cancer cells themselves
In vivo enhancement of metastatic growth:
Orthotopic implantation models reveal that MIF significantly increases tumor growth rate, volume, weight, and metastatic foci formation
Immunohistochemical analyses show that MIF promotes increased Ki-67 expression (proliferation marker) and decreased caspase-3 expression (apoptosis marker) in primary tumors
The relationship between MIF expression and estrogen in wound healing reveals a complex regulatory mechanism:
Age-associated estrogen regulation of MIF:
Inverse correlation between estrogen and MIF levels:
Clinical evidence in humans:
Studies comparing premenopausal females, postmenopausal females, and hormone replacement therapy (HRT)-treated postmenopausal females demonstrate that HRT (estrogen supplementation) reverses age-related increases in MIF levels
These findings indicate that estrogen therapy may improve wound healing outcomes by modulating MIF expression
Molecular mechanisms:
Estrogen appears to regulate MIF at the transcriptional level
Experiments with the MIF promoter (1-kb fragment derived from GenBank AF033192) cloned into reporter plasmids demonstrate estrogen-responsive elements
Both ERα and ERα lacking the AF-1 transactivation domain have been studied for their effects on MIF promoter activity
Cellular responses:
In vitro studies with human peripheral circulating monocytes show that estrogen treatment (at 10^-8 M) can modulate MIF expression
This regulation is particularly relevant in inflammatory contexts, such as after lipopolysaccharide (LPS) stimulation
The modulation of MIF by estrogen affects monocyte/macrophage functions critical for wound healing
Targeting MIF offers several promising therapeutic applications across multiple disease contexts:
Inflammatory and autoimmune diseases:
As a pro-inflammatory mediator, MIF has been implicated in the pathogenesis of severe sepsis, septic shock, acute respiratory distress syndrome, rheumatoid arthritis, glomerulonephritis, and inflammatory bowel diseases
Pharmacological inhibition of MIF could potentially reduce inflammation in these conditions
Small molecule inhibitors like ISO-1 and P425, which have shown efficacy in inhibiting MIF-induced cell migration in experimental models, represent potential therapeutic candidates
Cancer treatment approaches:
Given MIF's role in promoting tumor growth, metastasis, and EMT, anti-MIF strategies could target multiple aspects of cancer progression
Potential approaches include:
Small molecule inhibitors targeting MIF's enzymatic activity
Neutralizing antibodies against MIF
RNA interference to reduce MIF expression
Targeting downstream signaling pathways activated by MIF
Hepatocellular carcinoma (HCC):
MIF promoter polymorphisms (particularly rs755622) are associated with increased susceptibility to HCC and poorer prognosis
Stratifying patients based on MIF genotypes could help identify those who might benefit most from anti-MIF therapies
Patients with GC and CC genotypes, who express higher levels of MIF protein, might be prime candidates for such interventions
Wound healing applications:
The inverse relationship between estrogen and MIF levels suggests potential therapeutic applications for wound healing, particularly in populations with reduced estrogen levels (e.g., elderly individuals)
Options include:
Direct MIF inhibition to potentially improve wound healing in estrogen-deficient states
Combined estrogen and anti-MIF therapies for synergistic effects
Topical applications to avoid systemic effects of estrogen therapy
Biomarker potential:
MIF levels in peripheral blood could serve as biomarkers for disease progression, particularly in HCC where they correlate with poor prognosis
Monitoring MIF levels might help assess treatment efficacy
Genetic screening for MIF polymorphisms could help identify patients at higher risk for certain diseases, enabling earlier intervention
Macrophage Migration Inhibitory Factor (MIF) is a small, secreted protein that plays a crucial role in the immune system. It acts as a pro-inflammatory cytokine and an enzyme, influencing various immune responses. The recombinant form of MIF, particularly the human recombinant with a His tag at the C-terminus, is widely used in research to study its functions and potential therapeutic applications.
Human recombinant MIF is typically expressed in insect cells and purified to high levels of purity, often exceeding 98% as determined by SDS-PAGE and HPLC analyses . The recombinant protein is a 15 kDa molecule containing 124 amino acid residues, including an N-terminal His-tag . This His-tag facilitates purification and detection of the protein in various experimental setups.
MIF is known for its dual role as a cytokine and an enzyme. As a cytokine, MIF is involved in the regulation of immune responses. It is secreted by various cells, including macrophages and T cells, in response to inflammatory stimuli. MIF binds to receptors such as CD74 and CD44 on target cells, triggering the release of other pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α), interleukins (IL-1, IL-6, IL-8), and matrix metalloproteinases (MMPs) .
As an enzyme, MIF exhibits tautomerase activity, catalyzing the keto-enol isomerization of substrates like keto-phenylpyruvate and L-dopachrome . This enzymatic activity is dependent on its trimeric configuration and the presence of a free N-terminal proline residue .
MIF has been implicated in various diseases due to its pro-inflammatory properties. It plays a significant role in conditions such as sepsis, autoimmune diseases, and cancer. Elevated levels of MIF have been observed in patients with these conditions, making it a potential target for therapeutic interventions .
The human recombinant MIF with a His tag at the C-terminus is a valuable tool in research. It allows scientists to study the protein’s structure, function, and interactions in detail. The His tag aids in the purification process, ensuring that high-purity protein is available for experiments. Additionally, recombinant MIF is used in assays to investigate its role in immune responses and its potential as a therapeutic target .