Mif Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Target and Biological Significance

MIF (UniProt ID: P14174) is a 12.5 kDa protein encoded by the MIF gene (NCBI Gene ID: 4282) that functions as a:

  • Pro-inflammatory cytokine regulating innate immunity

  • Counter-regulator of glucocorticoid immunosuppression

  • Enzymatic catalyst (L-dopachrome isomerase/tautomerase)

Structurally, MIF forms a homotrimer with two antiparallel α-helices and a four-stranded β-sheet per monomer . Its interaction with receptors CD74 and CXCR4 drives inflammatory signaling, making it a therapeutic target in autoimmune diseases and cancer .

Antibody Development and Functional Characterization

Anti-MIF antibodies are generated through phage display libraries and hybridoma technology, targeting distinct epitopes:

Epitope Region (Amino Acids)Functional ImpactNeutralization Efficacy
50–68 (β-sheet domain)Blocks MIF-CD74 interaction27% of antibodies show neutralizing activity
86–102 (oxidoreductase motif)Inhibits tautomerase function32% of antibodies neutralize MIF
C-terminal (AA 3–111)Binds secreted MIFUsed in diagnostic assays

Key functional assays for validation:

  • Cell proliferation inhibition (IC₅₀: 10–100 nM)

  • Tautomerase activity reduction (≥50% at 1 μg/mL)

  • Glucocorticoid antagonism suppression (IL-6 reduction >25%)

Clinical-stage MIF inhibitors demonstrate disease-modifying potential:

AntibodyTargetClinical Trial PhaseOutcome
ImalumabMIF β-sheetPhase I (NCT01765790)26% disease stabilization in solid tumors
MilatuzumabCD74 receptorPhase II (NCT00504972)Orphan Drug status for myeloma
CXCR4 antagonistsMIF signalingPhase II (NCT02826486)Combinatorial immunotherapy trials

Mechanistic advantages:

  • Reduces DC MHC-II downregulation (70% reversal in vitro)

  • Inhibits tumor-associated macrophage infiltration (≥50% in murine models)

  • Blocks MIF-mediated PD-L1 upregulation

Technical Challenges and Innovations

Current limitations driving antibody engineering:

  • Epitope masking by MIF's trimeric structure (solved via β-sheet targeting)

  • Species cross-reactivity gaps (addressed by phage display maturation)

  • Short serum half-life (improved via Fc region modifications)

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we are able to ship the products within 1-3 business days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. We recommend reaching out to your local distributors for specific delivery details.
Synonyms
MifMacrophage migration inhibitory factor antibody; MIF antibody; EC 5.3.2.1 antibody; Delayed early response protein 6 antibody; DER6 antibody; Glycosylation-inhibiting factor antibody; GIF antibody; L-dopachrome isomerase antibody; L-dopachrome tautomerase antibody; EC 5.3.3.12 antibody; Phenylpyruvate tautomerase antibody
Target Names
Mif
Uniprot No.

Target Background

Function
Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that plays a pivotal role in the innate immune response to bacterial pathogens. Its expression at sites of inflammation suggests a crucial role as a mediator in regulating macrophage function during host defense. MIF counteracts the anti-inflammatory activity of glucocorticoids. It exhibits phenylpyruvate tautomerase and dopachrome tautomerase activity in vitro, although the physiological substrate remains unknown. The physiological relevance of tautomerase activity and its potential contribution to cytokine activity are currently unclear.
Gene References Into Functions
  1. MIF mediates LPS-induced cardiac dysfunction in murine cardiomyocytes, which was attenuated by MIF knockout. PMID: 29350381
  2. MIF attenuates oxygen-glucose deprivation-induced cochlear cells injury. MIF enhances Nrf2 and inhibits oxidative stress in cochlear cells. The enhanced Akt-Nrf2-HO-1 pathway may mediate cochlear protection by MIF. PMID: 29908183
  3. Evidence suggests that macrophage migration inhibitory factor (MIF) functions as a regulator of the NLR family pyrin domain containing 3 (NLRP3) inflammasome complex in macrophages. PMID: 29884801
  4. Research indicates that macrophage migration inhibitory factor directly engages in dengue NS1-induced glycocalyx degradation. Targeting MIF may present a potential therapeutic approach for preventing dengue-induced vascular leakage. PMID: 29702687
  5. Findings suggest a model in which MIF expression in the primary tumor dampens the anti-tumor immune response, thereby promoting tumor growth. PMID: 29864117
  6. MIF knockdown significantly exacerbates hearing loss in young mice. PMID: 28990052
  7. MIF mediates PAR4-induced bladder pain through urothelial HMGB1. PMID: 29263120
  8. Research reveals that high systemic levels of MIF contribute to the development of type 2 diabetes mellitus pathology. PMID: 28780379
  9. High MIF expression is associated with progressive multiple sclerosis. PMID: 28923927
  10. The absence of MIF leads to disturbances in systemic and hippocampal insulin sensitivity, which are potentially responsible for memory deficits and anxiety. This may occur through decreased PSA-NCAM-mediated neuroplasticity rather than through neurotrophic factors. PMID: 28919555
  11. Data indicate the functional role of the MIF-COX-p53 axis in inflammation and cancer at the genomic and proteomic levels in COX-2-ablated cells. PMID: 29247872
  12. Findings demonstrate that MIF regulates MCP-1 expression in hepatocytes of injured liver via CD74, CD44, and p38 MAPK in an autocrine manner. PMID: 27273604
  13. MIF is implicated in the pathogenesis of AF, possibly by down-regulating the protein and gene expression of Cx43 via ERK1/2 kinase activation. PMID: 28429502
  14. Endogenous MIF reduces the accumulation and toxicity of misfolded SOD1 in a mouse model of amyotrophic lateral sclerosis. PMID: 27551074
  15. Gene expression of MIF was 30-fold higher in the heart, compared to skeletal muscle, and protein expression of MIF was 3-fold higher in the heart compared to skeletal muscle. PMID: 27364992
  16. Renal tubular MIF is an endogenous renoprotective factor in progressive kidney diseases. PMID: 28801314
  17. Locally produced MIF in the inflammatory bone lytic site is involved in the chemoattraction of circulating CXCR4+ osteoclast precursor cells. PMID: 27082509
  18. MIF expression was induced in chondrocytes of tissue-engineered cartilage, and could exert a profound effect on chondrocytes by promoting cartilage maturation. MIF could also regulate the phenotype of surrounding macrophages, impairing the maturation of transplanted tissues. PMID: 28574571
  19. Pretreatment of P. aeruginosa with rMIF is associated with reduced bacterial killing by tobramycin. PMID: 28768722
  20. Loss of autophagy, by pharmacological inhibition or siRNA silencing of Atg5, enhances MIF secretion by monocytes and macrophages. PMID: 27163877
  21. CHD7 is an important factor in the proliferation and stemness maintenance of neural stem/progenitor cells. PMID: 27955690
  22. MIF-deficient mice have reduced Nippostrongylus brasiliensis burden and mounted an enhanced type 2 immune response, including increased Gata3 expression and interleukin-13 production in the mesenteric lymph nodes. PMID: 27049059
  23. Sertoli cells produce MIF under normal conditions. MIFR is expressed in GFRalpha1 and Sertoli cells. MIF induced spermatogonial cell migration. PMID: 27925200
  24. MIF-transgenic cells exhibited substantially decreased levels of p53 after hyperthermia treatment compared with WT and MIF-knockout cells. PMID: 27528627
  25. Research demonstrated that loss of keratinocyte-derived MIF leads to a loss of control of epithelial skin tumor formation in chemical skin carcinogenesis, highlighting an unexpected tumor-suppressive activity of MIF in murine skin. PMID: 27825106
  26. This study aimed to investigate the potential role of Macrophage migration inhibitory factor in osteoarthritis in human joint tissues and in vivo in mice with age-related and surgically induced osteoarthritis. PMID: 27564840
  27. MIF (macrophage migrating inhibitory factor), a potential pathogenic molecule in African trypanosomosis, was found to promote erythrophagocytosis, block extramedullary erythropoiesis and RBC maturation, and trigger hemodilution. PMID: 27632207
  28. Findings suggest that macrophage migration inhibitory factor regulates extramedullary erythropoiesis by inhibiting an overexpansion of splenic immature erythroid cells during chronic stress, indicating a novel role for this cytokine under chronic stress conditions. PMID: 27129368
  29. Findings suggest that Mif plays a role in the molecular mechanisms of macrophage and dendritic cell activation and drives T cell responses involved in the pathology of type 1 diabetes mellitus. PMID: 27699180
  30. MIF has a potential role in pathological angiogenesis of proliferative retinopathy. PMID: 28070752
  31. Genetic Mif deletion reduces the incidence and severity of oral carcinogenesis, by inhibiting the expression of chronic pro-inflammatory immune mediators. Thus, targeting MIF is a promising strategy for the prevention or therapy of oral cancer. PMID: 27164411
  32. MIF inhibits myoblast differentiation by affecting cell cycle progression, but does not affect proliferation. PMID: 26927414
  33. This paper demonstrates that the detrimental effect of MIF knockout was associated with accentuated loss in cardiac autophagy with aging. PMID: 26940544
  34. Research suggests that MIF promotes mCSC survival, proliferation, and endothelial differentiation through the activation of the PI3K/Akt/mTOR and AMPK signaling pathways. PMID: 27035848
  35. Posttranslational modification of MIF by S-nitrosation results in intracellular accumulation and protection from myocardial ischemia reperfusion injury. PMID: 26310191
  36. Data show that siRNA-induced macrophage migration inhibitory factor (MIF) reduction in murine mammary cancer line 4T1 and human breast cancer line MDA-MB-231 resulted in significant reduction of cell proliferation and increase of apoptosis. PMID: 26403072
  37. High expression levels of macrophage migration inhibitory factor sustain the innate immune responses of neonates. PMID: 26858459
  38. The deletion of the MIF gene led to reduced behavioural despair in mice of both sexes and IFN-gamma mRNA levels were reduced in the hippocampus of the MIF KO mice. PMID: 26338025
  39. In D-galactosamine-sensitized mice CP+Cu(II) increased the LPS-induced lethality from 54 to 100%, while administration of antibodies against MIF prevented the lethal effect. The enhancement by CP+Cu(II) of the pro-inflammatory signal of MIF is discussed. PMID: 26091949
  40. Data suggest that the MIF-Notch axis may play an important role in the pathogenesis of experimental autoimmune uveitis. PMID: 26400205
  41. The functional role of MIF in cell recruitment was investigated by a chemotaxis assay and by flow cytometry of labeled macrophages that were injected into Mif-/-and wildtype mice. PMID: 26348853
  42. These results implicate MIF in the pathogenesis of esophageal inflammation and suggest that targeting MIF might represent a novel therapy for EoE. PMID: 25712805
  43. Data suggest that macrophage migration inhibitory factor (MIF) inhibition could be a promising approach to the treatment of diabetes mellitus (DM)-associated atherosclerosis (AS). PMID: 25661015
  44. Bladder PAR activation elicits urothelial MIF release and urothelial MIF receptor signaling at least partly through CXCR4 to result in abdominal hypersensitivity without overt bladder inflammation. PMID: 26020638
  45. Transcription factor MEF2 and Zac1 mediate MIF-induced GLUT4 expression through CD74-dependent AMPK activation in cardiomyocytes. PMID: 26455966
  46. Blockade of CXCR7 suppressed MIF-mediated ERK- and zeta-chain-associated protein kinase (ZAP)-70 activation. PMID: 26139098
  47. Macrophage migration inhibitory factor is detrimental for survival and is associated with lung pathology, inflammatory cellular infiltration, and bacterial replication in a mouse model of pneumococcal pneumonia. PMID: 25943202
  48. Macrophage migration inhibitory factor may play an important role in recovery from acoustic trauma. PMID: 25853607
  49. Data indicate that MIF and CD74 facilitate RANKL-induced osteoclastogenesis, and suggest that MIF contributes directly to bone erosion, as well as inflammation, in rheumatoid arthritis. PMID: 25647268
  50. MIF was found to be a major platelet-derived chemotactic recruitment factor with clot-modulating properties and therefore might be relevant in inflammatory diseases such as atherosclerosis. PMID: 25561410

Show More

Hide All

Database Links

KEGG: mmu:17319

STRING: 10090.ENSMUSP00000041149

UniGene: Mm.2326

Protein Families
MIF family
Subcellular Location
Secreted. Cytoplasm.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : IP

Sample type: U138MG cells

Review: γH2AX associates with MIF: Chromatin fraction was isolated from U138MG cells 7 days post siAEBP1 transfection. γH2AX was immunoprecipitated using Protein A agarose beads and the eluate from the matrix was probed with anti-AIF, anti-γH2AX and anti-MIF antibodies.

Q&A

What is Macrophage Migration Inhibitory Factor (MIF) and why are anti-MIF antibodies important research tools?

MIF is a pro-inflammatory cytokine involved in the innate immune response to bacterial pathogens. The expression of MIF at sites of inflammation suggests its role as a mediator in regulating macrophage function in host defense . MIF counteracts the anti-inflammatory activity of glucocorticoids and has enzymatic activities including phenylpyruvate tautomerase and dopachrome tautomerase, though their physiological relevance remains unclear .

Anti-MIF antibodies are critical research tools that enable scientists to:

  • Detect and quantify MIF protein in various experimental settings

  • Neutralize MIF activity in functional assays

  • Study MIF's role in inflammatory and autoimmune conditions

  • Evaluate MIF as a potential therapeutic target

MIF has been implicated in numerous disease states including rheumatoid arthritis, sepsis, inflammatory bowel disease, and autoimmune disorders, making anti-MIF antibodies valuable for both basic research and translational medicine .

What are the different types of MIF antibodies available for research applications?

Researchers can choose from several types of MIF antibodies:

Antibody TypeDescriptionCommon ApplicationsConsiderations
PolyclonalRecognize multiple epitopes on MIFWB, IHC-P, ELISAHigher sensitivity, potential cross-reactivity
MonoclonalTarget a single epitope on MIFWB, IP, IF, IHCP, ELISAHigher specificity, consistent performance
RecombinantProduced using recombinant DNA technologyMultiple applicationsReduced batch-to-batch variability
NeutralizingBlock MIF biological activityFunctional assays, in vivo studiesSpecific for active regions of MIF
ConjugatedLinked to tags (HRP, fluorophores, etc.)Flow cytometry, direct detection assaysNo secondary antibody needed

For example, the MIF Antibody (D-2) is a mouse monoclonal IgM antibody that detects MIF in mouse, rat, and human samples through applications including Western blotting, immunoprecipitation, immunofluorescence, immunohistochemistry, and ELISA .

How should I select the optimal MIF antibody for my specific research application?

Selection of the appropriate MIF antibody depends on several critical factors:

  • Target species reactivity: Ensure the antibody recognizes MIF from your species of interest (human, mouse, rat, etc.)

  • Application compatibility: Verify the antibody is validated for your specific application (WB, IHC, IF, ELISA, etc.)

  • Epitope specificity: For certain applications, you may need antibodies targeting specific regions of MIF:

    • Antibodies binding the β-sheet structure (amino acids 50-68 or 86-102) have demonstrated neutralizing activity in functional assays

    • Structural vs. linear epitope recognition impacts functionality

  • Validation data: Review available validation data for the antibody in your application of interest

  • Clone selection: Different clones may have variable performance in specific applications or with certain sample types6

Based on published research, antibodies specific for the β-sheet structure of MIF that includes the oxidoreductase motif have shown potent inhibitory activity in both in vitro and in vivo studies .

What validation methods should I use to confirm the specificity of my MIF antibody?

Thorough validation is critical for generating reliable results with MIF antibodies. Consider these validation strategies:

  • Positive and negative control samples:

    • Use cell lines/tissues known to express or lack MIF

    • Include MIF knockout or knockdown controls when possible

  • Epitope mapping:

    • Determine if your antibody recognizes linear or structural epitopes

    • Use MIF-derived peptides spanning different regions of the protein to identify binding regions

  • Cross-reactivity assessment:

    • Test for reactivity with related proteins or other species

    • Verify lack of cross-reactivity between human MIF and parasite-produced MIF antibodies when working with infectious disease models

  • Functional validation:

    • For neutralizing antibodies, confirm inhibition of MIF-dependent activities:

      • Cell proliferation assays

      • Glucocorticoid-overriding activity (e.g., IL-6 secretion)

      • Tautomerase activity

  • Subcellular localization verification:

    • Confirm correct subcellular localization in immunofluorescence studies

    • Verify expected localization patterns (cytoplasmic and secreted for MIF)

Remember that detection of a specific band in Western blot does not guarantee antibody specificity in other applications like immunofluorescence .

How can I optimize my titration protocol for MIF antibodies in flow cytometry applications?

Antibody titration is critical for achieving optimal signal-to-noise ratio in flow cytometry. Follow these methodological steps for MIF antibody titration:

  • Preparation:

    • Use a cell population with known MIF expression

    • Prepare a single-cell suspension at consistent concentration (e.g., 1×10^6 cells/ml)6

    • Maintain constant time, temperature, and total volume across titration experiments

  • Titration setup:

    • Start with the manufacturer's recommended concentration

    • Create a serial dilution series (typically 2-fold)

    • Include both positive and negative controls for each dilution6

  • Analysis metrics:

    • Calculate separation index: (MFI positive - MFI negative) / (2 × SD of negative)6

    • Determine staining index: (Median positive - Median negative) / (2 × SD of negative)

    • Plot metrics against antibody concentration to identify optimal titer6

  • Optimal titer identification:

    • Select the concentration that provides:

      • Saturation of positive population (plateau in MFI)

      • Minimal spread/background in the negative population

      • Maximum separation between positive and negative populations6

  • Panel considerations:

    • Re-evaluate optimal titer in context of full panel due to fluorochrome interactions

    • Consider steric hindrance when multiple antibodies target proximal epitopes6

Remember that the optimal titer is typically found at the saturation phase of the curve, just before excess antibody leads to increased background6.

What are the key considerations for developing neutralizing anti-MIF antibody therapies?

Developing neutralizing MIF antibodies as potential therapeutics requires addressing several important factors:

  • Target epitope selection:

    • Antibodies binding the β-sheet structure (amino acids 50-68 or 86-102) demonstrate the most potent MIF inhibition

    • The β-sheet structure includes the MIF oxidoreductase motif, which appears crucial for activity

    • Antibodies targeting linear epitopes spanning amino acids 2-45 and 69-85, by contrast, show limited neutralizing activity

  • Cross-reactivity considerations:

    • Ensure specificity for target MIF (e.g., human MIF) without cross-reactivity to homologous proteins

    • Evaluate potential cross-reactivity with other species' MIF when developing for clinical use

    • Human and parasite MIF homologs share structural similarities but low sequence identity (28%), enabling development of specific antibodies

  • Functional validation methods:

    • Cell proliferation assays to assess inhibition of MIF-dependent proliferation

    • Evaluation of glucocorticoid-overriding activity (e.g., measuring IL-6 secretion)

    • Tautomerase activity assays to assess enzymatic inhibition

    • In vivo models of relevant diseases (e.g., sepsis, inflammatory conditions)

  • Antibody engineering approaches:

    • Consider antibody format (IgG subclass, Fab fragments, bispecifics)

    • Evaluate valency effects (1:1, 2:1, or 2:2 binding configurations)

    • Explore Fc engineering to enhance or reduce effector functions based on therapeutic goals

Recent studies demonstrate that combination therapy with neutralizing anti-MIF antibodies alongside standard treatments can provide superior outcomes compared to monotherapy, particularly in severe infectious diseases .

What are the common causes of non-specific binding with MIF antibodies and how can they be addressed?

Non-specific binding can significantly impact the quality of MIF antibody-based experiments. Here are common causes and solutions:

IssuePossible CausesSolutions
High background in immunoassaysInsufficient blockingUse BSA/FBS as blocking agents (typically 5-10%)
Optimize blocking time and concentration
Fc receptor bindingFc receptors on cells (especially myeloid cells) binding antibody Fc regionUse Fc receptor blocking reagents:
- Human: 10% homologous serum or commercial Fc block
- Mouse: anti-CD16/32
Direct dye binding to myeloid cellsSome fluorochromes directly bind monocytes/myeloid cellsUse TrueStain Monocyte blocker for flow cytometry applications
Fluorochrome aggregatesBrilliant Violet dyes can form aggregatesUse BV staining buffer
Centrifuge antibody vial (10,000 RPM for 3 min) before use
Excess antibody concentrationToo high antibody concentration causing non-specific bindingPerform proper antibody titration
Use the minimum concentration needed for specific detection
Dead cell bindingDead cells becoming sticky and autofluorescentInclude viability dye in staining protocols
Use amine-reactive (fixable) dyes for fixed samples
Use DNA-binding dyes (PI, 7-AAD) for live cell analysis

Always include appropriate negative controls in your experimental design to assess the level of non-specific binding .

How should I troubleshoot inconsistent results when using MIF antibodies across different experimental replicates?

Inconsistent results with MIF antibodies can stem from various sources. Follow this systematic troubleshooting approach:

  • Antibody storage and handling:

    • Verify proper storage conditions (temperature, avoid freeze-thaw cycles)

    • Check antibody expiration date

    • Confirm antibody hasn't been left at room temperature or exposed to light6

  • Sample preparation inconsistencies:

    • Standardize cell/tissue lysis protocols

    • Use consistent fixation and permeabilization methods

    • Filter samples to prevent aggregation (add EDTA 2-5mM, except when studying adhesion molecules)

    • Handle samples gently to prevent cell death

  • Protocol variations:

    • Maintain consistent incubation times and temperatures

    • Use the same buffers across experiments

    • Standardize washing steps6

  • Lot-to-lot antibody variability:

    • Consider lot testing before switching to a new antibody lot

    • Document lot numbers used in experiments

    • Consider using recombinant antibodies with reduced batch variability

  • Instrument variations (for flow cytometry):

    • Perform regular instrument quality control

    • Use standardized beads for fluorescence calibration

    • Maintain consistent instrument settings across experiments6

  • Biological variation:

    • Control for cell culture conditions (passage number, confluence)

    • Consider biological variability in primary samples

    • Include appropriate positive and negative controls in each experiment

For critical experiments, consider running all samples simultaneously rather than across multiple days to minimize technical variability.

How can MIF antibodies be utilized in therapeutic development for inflammatory and autoimmune diseases?

MIF antibodies show significant potential as therapeutic agents for various inflammatory and autoimmune conditions:

  • Target disease indications:

    • Rheumatoid arthritis: MIF high-expression alleles correlate with disease severity

    • Sepsis and septic shock: Anti-MIF antibodies demonstrate protective effects in animal models

    • Inflammatory bowel disease: Neutralizing MIF antibodies show beneficial effects in experimental models

    • Contact hypersensitivity: Antibodies binding specific epitopes exert protective effects

    • Systemic lupus erythematosus (SLE): MIF alleles may influence end-organ damage susceptibility

  • Therapeutic mechanisms:

    • Inhibition of MIF's pro-inflammatory cytokine activities

    • Prevention of MIF-induced overproduction of TNF-α, IL-1, and IFN-γ

    • Reduction of MIF-mediated tissue damage through proteinase induction

    • Restoration of glucocorticoid sensitivity in inflammatory conditions

  • Antibody development approaches:

    • Fully human antibodies minimize immunogenicity concerns

    • Structure-guided antibody design targeting the β-sheet region (aa 50-68 or 86-102)

    • Phage display selection for high-affinity binders

    • Combination therapy with standard treatments may provide superior outcomes

  • Pharmacogenomic considerations:

    • MIF alleles occur in balanced polymorphism maintained by selective pressure

    • Patient MIF genotype may influence response to anti-MIF therapy

    • High-expression MIF alleles may predict better response to MIF-targeting approaches

Recent clinical development includes humanized anti-MIF antibodies that may offer promise for autoimmune disorders treatment . For example, a first-generation fully human IgG1 anti-oxMIF monoclonal antibody (imalumab) has been investigated in clinical trials for various cancers .

What are the methodological considerations for using MIF antibodies in multiplex immunoassays and flow cytometry panels?

Incorporating MIF antibodies into multiplex assays requires careful planning and optimization:

For CyTOF or spectral cytometry applications, special consideration should be given to panel design, as these platforms have unique spectral considerations different from conventional flow cytometry .

How are technological advancements shaping the development of next-generation MIF antibodies?

Recent technological innovations are driving significant advancements in MIF antibody development:

  • Antibody engineering platforms:

    • Bispecific antibodies: Anti-oxMIF/CD3 bispecifics for targeted recruitment and activation of T cells against tumor cells

    • Fc-engineered antibodies with enhanced effector functions for improved therapeutic efficacy

    • Antibody fragments and alternative scaffolds for improved tissue penetration

    • Radioimmunoconjugates as companion diagnostics for oxMIF-positive tumors

  • Target refinement:

    • Oxidized MIF (oxMIF) emerging as a more specific target in certain diseases

    • Co-development of therapeutic antibodies alongside companion diagnostics to enable targeted treatment of patients with oxMIF-positive tumors

    • Identification of specific epitopes (β-sheet structures including the oxidoreductase motif) as critical for therapeutic efficacy

  • Advanced screening technologies:

    • Phage display libraries generating diverse panels of fully human anti-MIF antibodies

    • High-throughput functional screening assays to identify antibodies with desired activities:

      • Cell proliferation inhibition

      • Glucocorticoid-overriding activity blockade

      • Tautomerase activity inhibition

  • Combination approaches:

    • Antibiotic-antibody combinations showing superior outcomes in infectious disease models

    • Anti-MIF antibodies combined with standard-of-care treatments for enhanced efficacy

    • Potential for reduced tissue damage in severe infections through combination therapy

Ongoing development includes anti-oxMIF antibodies in fast-track development for chronic inflammatory diseases, highlighting the continued innovation in this field .

What are the emerging applications of MIF antibodies in studying disease pathogenesis?

MIF antibodies are enabling new insights into disease mechanisms across multiple fields:

  • Parasite-host interactions:

    • Studies revealing parasite-produced MIF (e.g., E. histolytica MIF) as a virulence factor

    • Neutralizing antibodies against parasite MIF reducing inflammation and tissue damage without affecting host MIF function

    • Demonstration that anti-parasite MIF antibodies can block pathogen-stimulated cytokine production by human cells

  • Genetic associations and personalized medicine:

    • MIF allele studies revealing balanced polymorphisms that influence disease susceptibility and severity

    • High-expression MIF alleles protecting against certain infections while increasing autoimmunity risk

    • Potential for pharmacogenomic approaches to MIF-directed therapies based on patient genotype

  • Tissue damage mechanisms:

    • Investigation of MIF's contribution to inflammatory tissue damage through:

      • Induction of proteinases

      • Enhancement of pro-inflammatory cytokine production

      • Impairment of intestinal barrier integrity

    • Development of models to evaluate MIF's role in severe disease states

  • Novel disease associations:

    • Investigation of MIF in diseases beyond traditional inflammatory conditions

    • Expanding understanding of MIF's role in cancer biology

    • Exploration of MIF as a biomarker for disease severity and progression

MIF antibodies with high specificity and well-characterized functional properties will be essential tools for advancing these research areas and translating findings into clinical applications .

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 2025 TheBiotek. All Rights Reserved.