
Macrophage migration inhibitory factor: Exploring physiological roles and comparing health benefits against oncogenic and autoimmune risks (Review)
- Authors:
- Published online on: July 18, 2025 https://doi.org/10.3892/ijmm.2025.5590
- Article Number: 149
-
Copyright: © Camacho Meza et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Macrophage migration inhibitory factor (MIF) was first identified by Bloom and Bennett (1) in 1966 as a soluble factor produced by sensitized lymphocytes in response to specific antigens, inhibiting macrophage migration in vitro. They proposed three potential mechanisms for this inhibition, including the release of a pharmacological agent or an antibody-like substance, laying the foundation for understanding the role of MIF in immune responses (1,2).
The cloning of human MIF cDNA in 1989 provided the first clear insights into its biological activities (3). By 1994, bioactive MIF protein and neutralizing antibodies were being produced, revealing macrophages as both a source and target of MIF and underscoring its role in systemic inflammation (4). In 1995, MIF was identified as a glucocorticoid-induced modulator of cytokine production that balances pro- and anti-inflammatory responses (5). Structural experiments in 1996 revealed the unique ligand-receptor interactions of MIF, distinguishing it from other cytokines (6).
Between 1996 and 2003, the tautomerase activity of MIF and its involvement in infectious diseases, autoimmunity, cancer and atherogenesis were established (7-9). In 1998, the discovery of a CXXC motif in MIF explained its thiol-protein oxidoreductase activity, while its enzymatic role in detoxifying oxidized catecholamines suggested a protective function in neural tissue (10,11). The association of MIF with rheumatoid arthritis (RA) also emerged, with its inhibition proposed as a potential therapeutic strategy (12,13).
In 2003, the circadian rhythm of MIF was found to counteract the immunosuppressive effects of cortisol, and CD74 was identified as its high-affinity receptor, elucidating its molecular interactions (14,15). By 2006, CD44 was shown to mediate MIF-induced signaling via ERK1/2 phosphorylation (16) and in 2007, MIF was revealed as a non-cognate ligand for CXC motif chemokine receptors (CXCRs), further expanding its role in immune regulation (17).
A subsequent study in 2012 linked MIF to autophagy under stress conditions, and in 2021, MIF was proposed to function as a 3′ nuclease, facilitating cancer cell evasion of replication stress (18,19). These discoveries collectively underscore the multifaceted roles of MIF in inflammation, immunity and disease, making it a critical focus of ongoing therapeutic research. A timeline illustrating this decades-long journey in MIF research is presented in Fig. 1.
![]() |
Figure 1Although MIF was discovered nearly 60 years ago, our understanding of its role in a wide range of biological processes has significantly advanced only in recent decades. MIF, macrophage migration inhibitory factor; LPS, lipopolysaccharide. |
Localization and secretion
Cellular localization of MIF
MIF is primarily localized within the cytoplasmic vesicles of cells across most tissues, although it can also be detected in the nucleus (20). This dual localization enables the rapid secretion of MIF without the need for new protein synthesis. When MIF is required for secretion, an N-terminal signal sequence facilitates its translocation into the endoplasmic reticulum and Golgi apparatus for conventional processing. Additionally, previous studies have identified exosomes as an alternative secretory pathway, further expanding our understanding of the mechanisms involved in MIF release (20-22). The expression and secretion of MIF are enhanced by diverse stimuli, including pathogen-associated molecules such as lipopolysaccharide (LPS) and exotoxins, inflammatory mediators such as TNF-α and thrombin, metabolic factors such as glucose and insulin, and environmental stressors such as hypoxia and ultraviolet B irradiation. These findings demonstrate the complex regulation of MIF under various physiological and pathological conditions (20-22).
Mechanisms of MIF secretion
In addition to the aforementioned canonical pathway, MIF can also be secreted via a non-canonical route; however, the precise molecular mechanisms underlying this alternative pathway remain incompletely understood (23). Current research suggests that MIF release is mediated through programmed cell death pathways, including necrosis, necroptosis and NLRP3 inflammasome-dependent pyroptosis (20-22). Additionally, components of the vesicular transport system, such as the general vesicular transport factor p115 and the ATP-binding cassette transporter ABCA1, appear to participate in MIF secretion. Once released, MIF can act in an autocrine manner on the producing cell or in a paracrine fashion on neighboring cells, amplifying its biological effects (20-22).
Expression in different tissues
Originally characterized as a T cell-derived cytokine (1,2) MIF is now known to be widely expressed across various cell types and tissues. As detailed in Table I, MIF is produced by immune cells (such monocytes, macrophages and T lymphocytes), structural cells (such as epithelial, endothelial and smooth muscle cells) and specialized cells in the nervous system and pituitary gland. MIF is also detectable in circulating blood components, including white blood cells, plasma, red blood cells and platelets (24-28). Tissue-specific expression pattern analysis has revealed that MIF is present in the basal layer of the epidermis and in keratinocytes, where its levels increase during inflammatory skin conditions (29). Pancreatic islet β-cells also produce MIF in a glucose-dependent manner, while insulin-target cells such as skeletal muscle and adipose tissue exhibit variable secretion rates depending on anatomical location (30).
![]() |
Table IMIF expression in different cell types. |
Pathological significance
The pathological consequences of MIF hypersecretion are well documented (16,25,29). For instance, in vitiligo, elevated serum MIF is correlated with disease severity and duration, promoting autoimmune-mediated melanocyte destruction through macrophage recruitment and sustained inflammation (29). Similarly, in gastric cancer, MIF expression progressively increases from gastritis to malignancy, contributing to tumorigenesis via mechanisms including angiogenesis and immune evasion (25). Beyond its role in disease, MIF participates in physiological inflammatory responses by upregulating adhesion molecules (such as E-selectin, intercellular adhesion molecule-1 and Vascular Cell Adhesion Molecule-1) and chemokines (such as IL-8 and MCP-1), thereby enhancing leukocyte recruitment to sites of inflammation (22,31-34). Notably, in healthy individuals, serum MIF concentrations typically range from 0.2 to 8.3 ng/ml. However, these levels are significantly elevated in the aforementioned diseases, as well as in a wide range of other pathological conditions, including autoimmune disorders, chronic inflammatory diseases and diverse malignancies, as illustrated in Table II (16,25,29,35-46).
![]() |
Table IIMIF expression in certain pathological conditions. |
Genetics
The first reports describing the sequence of both MIF cDNA and its corresponding gene emerged in the early 1990s. The D-dopachrome tautomerase (D-DT) gene is closely related to the MIF gene and is considered to have originated through the duplication of a common ancestral gene. In humans, there is only one MIF gene, which is located on chromosome 22 (22q11.23). This gene consists of three exons measuring 107, 172 and 66 base pairs, and two introns measuring 188 and 94 base pairs, respectively. Both the exonic structure and the DNA sequence of MIF are highly conserved across species. As a result, MIF displays the highest degree of amino acid sequence homology among human-mouse cytokine pairs. MIF contains CpG islands, which contribute to its consistent expression as a single ~0.8 kb mRNA transcript (3). CpG islands are regions rich in cytosine and guanine nucleotides linked by phosphate bonds, typically located near gene promoters, which play a critical role in transcriptional regulation by serving as binding sites for transcription factors and influencing chromatin structure. In the case of MIF, the presence of CpG islands in its promoter region helps maintain an open chromatin configuration, promoting transcriptional activity and ensuring stable, constitutive gene expression under both physiological and pathological conditions (22,31-34).
The regulation of MIF gene expression is influenced by two well-characterized polymorphic sites located in the promoter region. The first consists of a variable number of CATT repeats at position-794, typically ranging from 5 to 8 copies. The second is a single nucleotide polymorphism at position-173 (G>C). The presence of >5 CATT repeats and the-173C allele has been associated with increased susceptibility to, and the severity of, various inflammatory and autoimmune diseases such as RA and systemic lupus erythematosus (SLE) (32). These polymorphisms have also been associated with an elevated risk of developing prostate and gastric cancer (22,31-34).
Structure and enzymatic activities
MIF has a homotrimer structure composed of three identical subunits. Each monomer has an approximate molecular weight of 12.5 kDa and contains 114 amino acids. The assembled trimer has a three-fold rotational symmetry with a solvent-accessible channel at its core. Each monomer comprises two antiparallel α-helices and a four-stranded β-sheet. Based on structural experiments, the central channel may serve as a potential binding site for small molecules. Initially, the biological function of this binding capacity was hypothesized to involve ligands such as glutathione, gangliosides or dopachrome (47).
As aforementioned, MIF possesses tautomerase activity, specifically catalyzing the conversion of D-dopachrome to 5,6-dihydroxyindole-2-carboxylic acid (11). Further enzymatic analyses have demonstrated that MIF also acts as a phenylpyruvate tautomerase, with phenylpyruvate and p-hydroxyphenylpyruvate as substrates, and as a thiol-protein oxidoreductase (10). These multifaceted catalytic functions have led to its classification as a cytozyme, reflecting its dual role as a cytokine and enzyme. Research by Matsunaga et al (11) revealed that MIF can catalyze the conversion of 3,4-hydroxyphenyl amine chrome and norepinephrine chrome, both toxic quinone derivatives of catecholamine neurotransmitters, into indole dihydroxy derivatives. These products may serve as precursors for neuromelanin, suggesting that MIF plays a protective role in neural tissue by detoxifying reactive catecholamine breakdown products (48). Structural experiments have shown that the N-terminal Pro1 is essential for catalytic function; insertion of an Ala between Pro1 and Met2, or replacement of Pro1 with Ser or Gly, abolishes tautomerase activity (11).
The enzymatic activity of MIF has been shown to be essential for optimal signaling in inflammatory and tumorigenic pathways. Additionally, the C-terminal domain has been implicated not only in enzymatic regulation but also in maintaining the tertiary structure of the protein (48). Consequently, the C-terminal region has emerged as a potential target for allosteric inhibition of enzymatic function. Among small-molecule inhibitors, 4-iodo-6-phenylpyrimidine (4-IPP) has been described as a 'suicide substrate' irreversibly inhibiting the tautomerase activity of MIF; it has demonstrated greater potency than ISO-1, with 4-IPP being associated with improved survival outcomes in murine models of sepsis. This specificity suggests that inhibition of MIF's enzymatic activity may preferentially affect tumorigenic processes while sparing physiological inflammatory responses (6,11,48,49).
Receptors and signaling pathways
CD74
Structure and expression
CD74 was the first non-polymorphic protein shown to associate with polymorphic major histocompatibility complex (MHC) class II molecules (50). Structurally, it comprises an N-terminal cytosolic tail, a transmembrane domain and a luminal C-terminal domain. The CD74 gene, located on chromosome 5, encodes four isoforms (50). CD74 is primarily expressed by professional antigen-presenting cells such as dendritic cells, B cells and macrophages, but it is also found in endothelial, epithelial and some mesenchymal cells (21).
Function and interaction with MIF
Within the endoplasmic reticulum, CD74 assembles into trimers and functions as a chaperone for MHC class II molecules, which is why it is also known as the 'class II invariant chain' (50). CD74 is produced in excess to ensure availability for MIF binding, independent of MHC II expression. Leng et al (15) reported that CD74, synthesized using a coupled transcription-translation reticulocyte lysate system, binds to MIF in vitro. Notably, a 40-amino acid segment within the extracellular domain of CD74 (residues 109-149) was identified as essential for mediating this interaction. Each CD74 trimer is capable of binding three MIF molecules. D-DT, a homolog of MIF, also binds CD74, albeit with faster association and dissociation kinetics than MIF (34).
Some parasites, including Plasmodium, Entamoeba, Toxoplasma and Leishmania, express MIF homologs that may exploit the MIF-CD74 interaction to evade host immune responses (50). MIF binds to the CD74-CD44 receptor complex, mediating inflammatory cell recruitment through downstream CXCR2 and CXCR4 signaling. This MIF-receptor interaction also activates the anti-apoptotic gene Bcl-xL, enhancing B cell survival; however, the precise mechanisms remain under investigation (20,21).
Additionally, MIF, through its interaction with the CD74 receptor, plays a pivotal role in the regulation of various physiological processes. MIF facilitates cell survival by activating Syk tyrosine kinase and the PI3K/Akt signaling pathway, culminating in the intramembrane cleavage of CD74 and the release of its intracellular domain (CD74-ICD) (51-53). In macrophages, phosphorylation of CD74-ICD by protein kinase A, followed by CD44 recruitment, leads to the activation of Src-family kinases and downstream ERK1/2 signaling, thereby modulating cellular metabolism (54). MIF also influences cell adhesion via the intracellular MIF-binding protein, Jun activation domain-binding protein 1 (JAB1), which sustains MIF-induced ERK phosphorylation (55). Under hypoxic conditions, MIF enhances hypoxia-inducible factor 1α (HIF1α) expression through a CD74-dependent autocrine positive-feedback mechanism (56). Furthermore, MIF/CD74/CD44 signaling exerts an inhibitory effect on osteoclastogenesis by inducing Lyn phosphorylation, which attenuates the receptor activator of NF-κB ligand-mediated Gab2/c-Jun N-terminal kinase (JNK)-1/c-Jun and Syk/phospholipase Cγ signaling pathways, ultimately suppressing the activation of nuclear factor of activated T cells 1 (57). These mechanisms are listed in Table III.
![]() |
Table IIIRoles of MIF/CD74 in physiological processes. |
CD74 in inflammation and signaling
CD74 plays an important role in tissue repair associated with inflammation, particularly in conditions such as inflammatory bowel disease, where it protects epithelial cells from oxidative stress in experimental models (50). MIF binding to CD74 activates the ERK1/2 pathway, a member of the mitogen-activated protein kinase (MAPK) family, which regulates cell proliferation, differentiation and stress responses (55,58). This interaction also enhances arachidonic acid metabolism, prostaglandin synthesis and Toll-like receptor expression via Ets-family transcription factors (59). Table IV illustrates additional downstream pathways activated by MIF-CD74 binding include PI3K/Akt, NF-κB and AMP-activated protein kinase (AMPK), all of which contribute to cellular survival, proliferation and metabolic regulation (28,50,58,60-65).
![]() |
Table IVCD74-mediated tissue repair. |
Regulation of CD74
CD74 expression is upregulated by interferon-γ (IFN-γ), a key mediator of both innate and adaptive immunity (50). CD74 also exists in a soluble form, generated through proteolytic shedding of its ectodomain, which reduces the availability of the membrane-bound receptor for ligand interaction (50). Interactions between MIF and CD74 are further modulated by proteins such as ribosomal protein S19 and JAB1, both of which act as competitive inhibitors of MIF binding (50).
CD44
Structure and function
CD44 is a key co-receptor in MIF signaling and plays important roles in cellular adhesion, migration, lymphocyte activation and angiogenesis. CD44 primarily functions through extracellular matrix (ECM) adhesion mechanisms (21,66). The CD44 gene, located on chromosome 11p13, contains 19 exons in humans, while mice possess an additional variant exon (v1). CD44 is broadly expressed in lymphocytes, fibroblasts and smooth muscle cells.
CD44 in disease and MIF signaling
CD44 has been studied in cancer due to the involvement of its splice variants in tumor progression (67). The extracellular domain of CD44 can bind to MIF, thereby modulating downstream signaling cascades, as illustrated in Fig. 2 (67). The splice variants v3 and v6 of CD44 are particularly elevated in T cells from patients with SLE and in fibroblast-like synoviocytes from patients with RA, contributing to enhanced invasiveness and inflammation (22). For MIF signal transduction to occur, CD74 and CD44 must form a cis-complex on the plasma membrane (68). This complex triggers the activation of Src-family tyrosine kinases and subsequent signaling through the PI3K/Akt and AMPK pathways (21). Binding of D-DT to the CD74/CD44 complex can also activate the ERK1/2 pathway (67). Beyond inflammation and cancer, CD44 has been implicated in metabolic diseases such as insulin resistance, likely due to its interactions with hyaluronan and modulation of MIF-related signaling (67).
![]() |
Figure 2MIF signaling pathway illustrates how this multifunctional molecule links immune response, inflammation and oncogenic processes. CXCR2/4/7, C-X-C motif chemokine receptor 2/4/7; TLR, Toll-like receptor; ABCA1, ATP binding cassette subfamily A member 1; ERK1/2, extracellular signal-regulated kinase 1/2; PLA2, phospholipase A2; FoxO3a, forkhead box O3a; JNK, c-Jun N-terminal kinase; GILZ, glucocorticoid-induced leucine zipper; COX2, cyclooxygenase-2; MKP-1, MAP kinase phosphatase 1; PGE2, prostaglandin E2; BAD, Bcl-2-associated agonist of cell death; BAX, Bcl-2-associated X protein; NFAT, nuclear factor of activated T cells; AP-1, activator protein 1; IFN-γ, interferon γ; ICAM, intercellular adhesion molecule; VCAM, vascular cell adhesion molecule; iNOS, inducible nitric oxide synthase; MMPs, matrix metalloproteinases; TXNIP, thioredoxin-interacting protein; Mdm2, mouse double minute 2 homolog. |
CXCRs: CXCR2, CXCR4 and CXCR7. MIF directly binds to three CXCRs, CXCR2, CXCR4 and CXCR7, each with distinct expression profiles. CXCR2 is predominantly expressed on neutrophils and endothelial cells, while CXCR4 and CXCR7 have broader distributions, including hematopoietic, endothelial and neuronal cells (22). Although MIF shares low sequence homology with natural ligands, such as CXC motif chemokine ligand (CXCL8) and SDF-1 (CXCL12), it contains specific motifs that enable receptor binding upon protein folding. Notably, D-DT does not bind CXCR2 (34). The prototypical function of MIF, inhibition of macrophage migration, is mediated by CD74-dependent recruitment of inflammatory cells through CXCR2 and CXCR4 (22,32,33,43,50). MIF also promotes monocyte adhesion and transendothelial migration, involving chemokines such as CXCL1 and CXCL8 (33).
CXCR-mediated signaling and immune modulation
MIF recruits myeloid-derived suppressor cells (MDSCs) via CXCL2-CXCR2 signaling, activating the MAPK and NF-κB pathways (31). However, MIF does not always mimic natural chemokine signaling. For example, it fails to activate ERK1/2 in platelets that lack CD74, indicating that CD74 is essential for CXCR2/4-mediated MIF signaling (21). Neutralization of CD74 abrogates signal transduction, reinforcing its critical role (21). When MIF binds to CD74 in complex with CXCR2 or CXCR4, it activates ERK1/2 and Akt through Gαi-coupled signaling. This complex may undergo internalization, potentially exposing these receptors on the surface of various tumors suggests that MIF-mediated effects are highly context-dependent (68).
CXCR7-specific roles
CXCR7 differs functionally from CXCR2 and CXCR4. Instead of initiating transient G-protein-coupled signaling, it shifts ERK1/2 activation toward a sustained β-arrestin-2-mediated pathway. Although CXCR7 was initially considered an atypical chemokine receptor, it is now recognized as a direct non-canonical receptor for MIF. MIF binding to CXCR7 can occur independently or in complex with CD74 and/or CXCR4, potentially modulating β-arrestin signaling and influencing cell migration and survival (20,21). CXCR7 has also been implicated in B cell trafficking and shown to activate ZAP-70, suggesting broader roles in immune regulation. These findings indicate that MIF may exert context-dependent effects through its interaction with CXCR7, distinct from classical chemokine pathways (21,22).
Roles of MIF in health
As aforementioned, MIF is a multifunctional protein that plays a crucial role in maintaining physiological homeostasis. MIF regulates immune responses by enhancing macrophage activity, promoting the production of pro-inflammatory cytokines, including TNF-α, IFN-γ, IL-1β, IL-2, IL-6 and IL-8, and facilitating pathogen clearance through the induction of nitric oxide (NO), cyclooxygenase-2 (COX-2) and efficient antigen presentation (69-71). MIF also modulates cellular proliferation and differentiation by activating key signaling pathways such as p44/42 ERK MAPK and cytosolic phospholipase A2. These pathways drive the production of inflammatory mediators (such as prostaglandins and arachidonic acid) and activate JNK/stress-activated protein kinase, which further amplify TNF-α signaling (22).
The broad physiological functions of MIF, detailed in Table V, also include organ protection. For example, MIF attenuates renal ischemia-reperfusion injury through its interaction with CD74 (59) and maintains cardiac redox homeostasis via S-nitrosylation at Cys81, reducing oxidative stress and apoptosis while activating the AMPK and ERK pathways (10,63,72-74). MIF also exerts neuroprotective effects, including the suppression of inflammation and apoptosis in Alzheimer's and Parkinson's disease, stimulation of autophagy and the rescue of neurons from superoxide dismutase 1-induced death (11,60,75-77). MIF contributes to tissue repair by promoting fibroblast and keratinocyte recruitment, enhancing angiogenesis and accelerating nerve regeneration through p53 suppression (78,79). In corneal and intestinal wounds, MIF activates Akt/ERK signaling to promote epithelial healing (80-82) Moreover, MIF plays a role in metabolic regulation, modulating insulin secretion via regulation of pancreatic and duodenal homeobox-1 and increased production of fructose-2,6-bisphosphate (30,83,84). This range of functions underscores the therapeutic potential of MIF in a variety of medical fields, including immunology, neurology, cardiology and metabolic disorders.
![]() |
Table VBeneficial effects of MIF in health. |
Roles of MIF in disease
MIF is widely recognized as a pivotal mediator in the pathogenesis of various inflammatory and immune-related diseases. MIF plays a central role in multiple pathological conditions, ranging from acute systemic inflammation to chronic autoimmune disorders and malignancies. For instance, in sepsis, MIF contributes to excessive and dysregulated inflammatory responses, exacerbating disease severity (85). In RA, MIF has been implicated in joint inflammation, synovial hyperplasia and cartilage degradation (86). MIF also plays a pathogenic role in diabetes mellitus, where it contributes to insulin resistance and pancreatic β-cell dysfunction (87). In the context of cancer, MIF promotes tumor progression, angiogenesis and immune evasion, underscoring its potential as a therapeutic target in solid tumors and other malignancies (33). The involvement of MIF in acute respiratory distress syndrome highlights its contribution to pulmonary inflammation and epithelial injury (88). Similarly, in multiple sclerosis (MS), MIF has been linked to neuroinflammation and disease exacerbation, further emphasizing its immunomodulatory role in neurological autoimmune disorders (89). MIF is also implicated in systemic autoimmune diseases such as SLE (90) and psoriasis (91), where it regulates immune cell activation and the secretion of pro-inflammatory cytokines. In dermatological conditions such as alopecia areata (AA), MIF may influence immune-mediated hair follicle destruction (92). In infectious diseases such as dengue, MIF contributes to the amplification of the inflammatory response and is associated with increased disease severity (22,93).
The pathogenic effects of MIF are mediated through several key mechanisms. One of the primary pathways involves the induction of inflammatory mediators, including TNF-α, NO, interleukins (such as IL-1, IL-6 and IL-8) and COX, all central components of the inflammatory cascade. MIF also modulates the response to LPS by acting in an autocrine manner to upregulate Toll-like receptor 4 and its co-receptor myeloid differentiation factor 2, thereby intensifying the immune response to bacterial endotoxins (22). Collectively, these findings suggest that MIF serves as a crucial link between innate immunity and chronic inflammation, reinforcing its potential as a therapeutic target in the treatment of diseases characterized by excessive immune activation and inflammatory damage.
MIF and cancer
Tumor microenvironment (TME)
The TME refers to the cellular and molecular milieu surrounding a tumor within the host's biological system. The TME is composed of vascular structures, immune and inflammatory cells (often bone marrow-derived), fibroblasts, ECM and the dynamic interactions among these components within the lymphatic and circulatory systems (94,95). The TME contributes significantly to tumor heterogeneity, progression and drug resistance. Non-malignant cells within the TME often play pro-tumorigenic roles throughout carcinogenesis, supporting uncontrolled cellular proliferation (95). For tumor development and metastasis to occur, cancer cells must acquire the ability to migrate, degrade the ECM, survive in circulation and colonize distant tissues. According to Mantovani et al (96), tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, supporting tumor progression by facilitating intravasation and protecting tumor cells from immune attack (94,96).
The TME is typically hypoxic, promoting the expression of MIF and other pro-inflammatory cytokines (96) (Fig. 3). Chronic inflammation, particularly mediated by MIF, promotes angiogenesis, resistance to apoptosis and tumor growth, which are hallmarks of malignancy (97-101). MIF contributes directly to these processes by activating key oncogenic pathways such as NF-κB, which acts as both a tumor initiator and promoter, and by inducing COX-2, which facilitates chemotherapy resistance. MIF also enhances STAT3 signaling, impairing dendritic cell function and allowing tumor immune evasion. Additionally, hypoxia and aberrant vasculature reinforce acidosis within the TME, which, in conjunction with MIF activity, drives malignant transformation and sustains the activation of HIF1α (Fig. 3) (96,102).
![]() |
Figure 3Numerous complex interactions occur between immune cells and cancer cells within the tumor microenvironment. This schematic provides a simplified overview of these interactions, with emphasis on the direct pro-tumorigenic effects of MIF. MIF contributes to immune evasion, chronic inflammation and tumor progression through its influence on cytokine production, immune cell polarization and signaling pathway activation. MIF, macrophage migration inhibitory factor; NK cell, natural killer cell; ILC, innate lymphoid cell; DC, dendritic cell; MDSC, myeloid-derived suppressor cell; CTL, cytotoxic T lymphocyte; TGF-β, transforming growth factor-β; IL, interleukin; IFN-γ, interferon-γ; TNF-α, tumor necrosis factor-α; COX-2, cyclooxygenase-2; PD-1, programmed death-1; PD-L1, programmed death ligand-1; CXCL12/14, C-X-C motif chemokine ligand 12/14. |
RAS family mutations are among the most frequent in human cancer (96). The RAS/RAF/MEK/ERK pathway triggers the production of pro-tumor cytokines and chemokines (96). The interaction between NF-κB and H1F1α leads to the upregulation of TNF-α and CXCR4, the latter being crucial for metastasis and a positive feedback loop for MIF expression (Fig. 3) (103). MIF, when secreted by cancer-associated fibroblasts (CAFs), promotes tumor cell activation by suppressing IFN signaling and inhibiting p53-dependent apoptosis (94); it also increases IL-6 production and amplifies tumor-promoting signals via the CXCR4/MIF/IL-6 loop (103). MIF also contributes to the polarization of macrophages toward a pro-inflammatory M1-like profile, yet its ultimate effect depends on the NF-κB mediated balance between proand antitumor functions (104). Thus, targeting MIF, its receptors (such as CD74 and CXCR4) and downstream mediators is necessary to disrupt the self-amplifying oncogenic loop (105). In parallel, strategies that re-educate tumor-promoting macrophages by modulating NF-κB could enhance antitumor immunity (Fig. 3) (94,106).
Quantifying MIF may have prognostic utility. A 2019 study identified elevated MIF and IL-17A levels as risk factors for breast cancer (BC) in the Mexican population (107). Novel therapies such as HSP90 inhibitors suppress MIF production in colorectal cancer (CRC) epithelial cells (108). Targeting MIF has shown promise in reducing the proliferation, angiogenesis and aggressiveness of cancer including triple-negative BC (TNBC), genitourinary cancer and pancreatic cancer (103,104,109,110). Additionally, combining autophagy inhibition with MIF targeting has been proposed for improved outcomes in TNBC (109).
BC
In BC, MIF contributes to carcinogenesis through upregulated pathways involved in proliferation, cell survival and growth. Among the molecular subtypes, high MIF levels are particularly associated with aggressive phenotypes, including the Luminal B, HER2-positive and TNBC subtypes (107). In TNBC, CD74 is strongly upregulated, leading to activation of the PI3K/Akt and MAPK pathways, which promote proliferation and survival. Co-expression of MIF and CD74 is also correlated with enhanced vascularity and elevated proangiogenic CXC chemokines, reinforcing the role of MIF in tumor aggressiveness and progression (63).
Beyond progression, MIF is linked to poor prognosis through its regulation of COX-2 expression and its facilitation of metastasis. Hypoxia upregulates MIF gene expression, further enhancing proliferation via PI3K/Akt activation. Moreover, HER2 upregulation induces heat-shock factor 1, which upregulates HSP90, a stabilizer of MIF (111). Collectively, these mechanisms contribute to uncontrolled tumor cell proliferation and highlight the value of MIF as a therapeutic and prognostic biomarker in BC.
Lung cancer
Similar to its role in BC, MIF significantly contributes to the progression of lung cancer, which remains the leading cause of cancer-related death in men and the second leading cause in women globally (112). Lung cancer is broadly classified into two main histological types: Non-small cell lung cancer (NSCLC) and small cell lung cancer (112). NSCLC represents 80-85% of all lung cancer cases, with over half of patients diagnosed at the advanced stages (112,113). MIF has a diagnostic and prognostic role by distinguishing normal lung tissues from NSCLC samples as well as correlating with elevated VEGF levels and increased microvessel density in tumors (114,115). Functionally, MIF promotes proliferation and inhibits apoptosis in NSCLC cell lines such as H460 and A549 (113).
At the molecular level, MIF is negatively regulated by microRNA-146a, a microRNA whose upregulation leads to reduced proliferation and increased apoptosis in lung cancer cells (115). Additionally, the CXCR4-upregulated MIF/IL-6 axis has been identified as a key driver of tumor-fibroblast crosstalk and NSCLC progression, suggesting therapeutic potential for CXCR4 inhibitors and MIF antagonists (103,113). MIF has also been associated with chemoresistance. Specifically, in cisplatin-resistant A549 and H460 cells, MIF expression is correlated with enhanced stemness and activation of the Src/CD155/MIF pathway, as well as upregulation of Notch1 and β-catenin. Inhibition of Src signaling has been shown to reduce these markers, supporting the strategy of combining Src inhibitors with chemotherapy in resistant NSCLC cases (116).
MIF also plays a key role in lung cancer metastasis, particularly to the brain, which occurs in up to 60% of NSCLC cases (117). Transcriptomic analysis by Liu et al (118) revealed high MIF expression in brain metastases. Additionally, blocking the MIF/CD74 axis enhances radiotherapy efficacy by shifting microglia toward the M1 antitumor phenotype, emphasizing the therapeutic value of targeting MIF in advanced NSCLC (117). In terms of therapeutic interventions, SCD-19, a small-molecule inhibitor that targets the tautomerase site of MIF, has demonstrated antitumor efficacy in a Lewis lung carcinoma model (119). Similarly, MD13, a PROTAC-based compound developed in China, induces MIF degradation and suppresses the MAPK pathway, inhibiting lung cancer cell growth (118).
CRC
In CRC, MIF contributes to inflammation-driven tumorigenesis and metastasis. The role of MIF in CRC parallels that observed in breast and lung cancer, reinforcing its broad relevance in oncology (120). One of the main mechanisms in the pathophysiology of this cancer involves the upregulation of CXCR4, a known MIF receptor, which enhances metastatic potential (121). Notably, CXCR4 is expressed in multiple cancer types, including breast, prostate and ovarian cancer, as well as immune cells such as B and T lymphocytes (121). Through interaction with CXCL12 and MIF, CXCR4 activates a variety of signaling cascades, including G protein, ERK, JNK and JAK/STAT pathways, that support tumor growth and spread (121,122). The CXCR4/MIF axis has thus emerged as a significant driver of CRC metastasis and a promising therapeutic target (121,122).
Elevated MIF expression has been detected in patients with CRC and is correlated with advanced disease stages (123). Moreover, the MIF gene-173G/C polymorphism has been associated with increased MIF protein expression and higher CRC susceptibility. Within the colonic microenvironment, elevated MIF may also influence intestinal cell kinetics and amplify inflammatory responses, creating conditions conductive to malignant transformation (120). These findings collectively highlight the importance of further research into MIF as both a biomarker and a therapeutic target in CRC.
MIF and autoimmunity
Autoimmunity refers to a spectrum of diseases characterized by inappropriate immune responses against self-antigens, leading to tissue damage and chronic inflammation. These conditions arise from a dysregulated adaptive immune response directed at specific anatomical structures (124). In recent years, autoimmune diseases have been increasingly recognized as a global issue due to their chronic nature, substantial healthcare costs and high prevalence among working-age individuals; nearly 4% of the world's population is affected by one of >80 different autoimmune diseases. Notably, women are affected three times more frequently than men, prompting the National Institutes of Health to classify autoimmunity as a major health concern in women (124-127).
Autoimmune pathologies are heterogeneous in localization and may become systemic. Experimental evidence links the onset of autoimmunity to abnormalities in self-antigen presentation. A dysregulated innate immune response can also trigger autoimmunity (124). Despite the wide variety of conditions, all known autoimmune diseases are thought to follow a sequence of initiation, propagation and resolution. Clinical manifestations generally begin during the propagation phase, which is characterized by progressive inflammation and tissue damage (124,128).
It is hypothesized that multiple polymorphisms may be required to disrupt regulatory mechanisms or lower threshold for lymphocyte activation, and that epigenetics plays an important role in the development of such pathologies (128,129). As previously discussed, MIF serves a crucial role in inflammation by promoting mediators such as TNF-α, IL-1, IL-6, IL-8, IL-12, IFNγ, COX-2, NO and matrix metalloproteinases. MIF also affects lymphocyte activation by supporting macrophage survival and modulating their function through the inhibition of p53 (129). In B cells, MIF binding to CD74 triggers a signaling cascade that activates NF-κB/p65/RelA homodimer and its coactivator, TAFII105, which regulate the transcription of genes controlling lymphocyte survival and proliferation (124,129). These processes contribute to a self-sustaining inflammatory loop (128). Autoimmunity propagation is considered to be related to an increased ratio of effector to regulatory T cells (Tregs). Notably (and paradoxically), the inflamed skin of patients with psoriasis does not show such a shift in ratio; however, Tregs appear to function abnormally and produce increased amounts of IL-17 (128).
Infections remain a major suspected trigger of autoimmunity (125). A combination of microbial exposure and genetic defects may result in a breakdown of immune tolerance and homeostasis. Some bacterial strains have been shown to behave differently depending on the in vivo or in vitro context (130).
SLE
High-expression MIF alleles have been identified as a risk factor for the development of SLE (35,131-133). Recent studies have focused on the role of MIF in glomerulonephritis, especially lupus nephritis (LN) (35,131-133). The relationship between MIF and active LN appears to be complex and may involve interactions with other molecules such as adiponectin or resistin. The presence of MIF in urine may reflect not only renal excretion but also local production by tubular epithelial cells or infiltrating leukocytes (133,134). MIF is considered a contributor to both disease activity and the cumulative damage associated with SLE.
Zhou et al (35) demonstrated that CD74, the receptor for MIF, is upregulated in kidney epithelial cells and antigen-presenting cells, and that this upregulation can be induced by IL-6 or IFN-γ in vitro, highlighting its potential role in the autoimmune processes of SLE. SLE progression often requires increasing steroid treatment, which appears to be correlated with type I IFN activity. IFN can induce MIF expression in a time- and dose-dependent manner (132,135). Beltrán-Ramírez et al (135) proposed that MIF may increase the expression of P-glycoprotein by upregulating inflammatory cytokines such as TNF-α and IL-6. A broader cytokine panel, incorporating temporal expression, has been suggested for defining additional SLE subsets (136).
While ISO-1, the prototypical MIF inhibitor, is not considered a viable therapeutic option due to its low potency, alternative strategies have been explored for SLE (137). Artesunate (ART), an antimalarial drug, has been shown to counteract the effects of type I IFNs by inhibiting STAT1 phosphorylation, resulting in stabilization or even regression of atherosclerotic plaques in experimental models. These findings point to the IFN/MIF axis as a novel target in SLE-associated atherosclerosis (132). Additionally, IMMU-115, a MIF-targeted therapy, has shown acceptable toxicity and preliminary efficacy in patients with SLE; it reduces LN symptoms, with disease suppression maintained for up to 24 weeks following the initial dose (138).
Psoriatic arthritis (PsA)
The role of MIF extends beyond SLE, playing a notable role in other autoimmune diseases such as PsA. PsA is a chronic inflammatory autoimmune disease marked by synovitis, enthesitis and aberrant bone remodeling, driven by dysregulated cytokine networks. Central to its pathogenesis is the IL-23/Th17 axis, with key cytokines such as IL-17, IL-22 and TNF-α playing pivotal roles in promoting inflammation, osteoclastogenesis and joint damage (139-143). Genetic predisposition, particularly involving HLA class I alleles such as HLA-B*27, and environmental triggers such as trauma and dysbiosis further contribute to the disease by amplifying aberrant immune responses (142,143).
MIF has emerged as a significant contributor to PsA pathogenesis. Secreted by macrophages, synovial fibroblasts and keratinocytes, MIF promotes synovial inflammation and joint destruction by enhancing the production of TNF-α, IL-17 and other cytokines, thereby sustaining the inflammatory loop and facilitating osteoclast (143,144). Compared with healthy controls, elevated serum and tissue levels of MIF have been documented in patients with PsA, supporting its involvement in disease pathology (140,145). Furthermore, genetic studies have implicated MIF promoter polymorphisms in the susceptibility to PsA. Notably, the-173 G>C variant, particularly the-173C allele and G/C genotype, has been associated with increased PsA risk in various populations, including Mexican Mestizos and British Caucasians (140,141,146) The CATT5/-173*C haplotype was also linked to psoriasis susceptibility in North Indian cohorts (145), reinforcing the relevance of MIF gene variants in psoriatic disease.
In psoriatic skin, MIF expression is upregulated in keratinocytes and endothelial cells, suggesting a broader role in the inflammatory processes of psoriatic disease beyond the joints (144). Experimental models further support this notion: MIF-deficient mice exhibit reduced keratinocyte hyperproliferation and inflammatory cell infiltration in psoriasiform dermatitis, indicating a potential mechanistic role for MIF in skin pathology (91). Despite these insights, additional studies are needed to fully delineate the multifaceted role of MIF in PsA and its potential as a biomarker or therapeutic target.
MS
Like PsA and SLE, MS is another autoimmune disorder in which MIF plays a crucial role. MS is the most common non-traumatic disabling disease affecting young adults (147). While its underlying cause remains uncertain, it is described as a chronic, immune-mediated demyelinating disease of the central nervous system (CNS) (89). MS is commonly viewed as a two-stage phenomenon, with early inflammatory processes driving the relapsing-remitting form and later neurodegeneration contributing to a non-relapsing progressive phase (147). During neuroinflammation, macrophages and microglia are considered significant sources of MIF in the CNS. Increased MIF levels have been found in patients with progressive MS, which are correlated with disease severity and progression (147).
A 2019 study by Cavalli et al (148) suggested that upregulation of the MIF cytokine family signature may occur in CD4+ T cells in patients with clinically isolated syndrome, potentially contributing to MS pathogenesis and serving as an effective biomarker and therapeutic target. The expression of MIF, CXCR4 and CD74 appears to be tightly regulated via negative feedback mechanisms (149). Similarly, Guan et al (150) demonstrated using a murine model of experimental autoimmune encephalomyelitis (a model of human MS) that inhibiting the MIF-CD74 interaction suppresses microglial M1 polarization and induces an anti-inflammatory response. B cell arrest in early maturation stages (implicated in MS onset) has been partially associated with MIF downregulation, which correlates with decreased CD74 and increased CXCR4 expression (89,149,151,152). Upregulation of CXCR4 suppresses Fas, reducing the clearance of autoreactive immune cells and allowing naive B cells to escape immune tolerance (89,149,152).
The relationship between the MIF-173G>C polymorphism and MS remains under investigation. Current evidence suggest no association with disease susceptibility in the Turkish population (129). However, in the Mexican Mestizo male population, this polymorphism may act as a male-specific modifier, influencing disease severity and progression (153).
Novel therapeutic strategies targeting MIF continue to emerge for MS. For instance, Dra1 constructs strongly inhibit the activation and recruitment of brain-infiltrating T cells and CD11+CD45high myeloid cells, which express elevated levels of CD74 following CNS damage. These constructs antagonize CD74 with high affinity, resulting in diminished MIF signaling. This treatment approach also shows potential in methamphetamine addiction and traumatic brain injury (154). Additionally, iguratimod (IGU), an anti-rheumatic drug, exhibits selective MIF inhibition both in vitro and in vivo, and demonstrates additive effects with glucocorticoids in autoimmune encephalitis models (155). Another promising drug is ibudilast, a MIF and phosphodiesterase inhibitor, which has shown efficacy in slowing brain atrophy in progressive MS (156). It is worth noting that MIF levels are not useful for distinguishing responders from non-responders to glucocorticoid treatment in patients with acute optic neuritis, as they reflect only ongoing inflammation seen in long-term MS progression (157).
AA
As a more organ-specific autoimmune condition, AA exemplifies how the immunomodulatory role of MIF impacts localized immune privilege. AA is characterized by non-scaring hair loss, ranging from bald patches to total scalp involvement. The disease emerges when immune privilege is lost in hair follicles, and MIF has been implicated in this process due to its ability to suppress natural killer cell activity (92,158,159). Genetics, emotional stress and autoimmunity all play an important role in the pathogenesis of AA (158).
Based on these findings, Eldesouky et al (158), investigated the involvement of MIF in AA and vitiligo using a small cohort comprising 22 patients with AA, 20 patients with vitiligo and 20 healthy controls. The results showed that MIF levels were significantly elevated in both AA (8.477±4.1761 ng/ml) and vitiligo vulgaris (3.930±2.7071 ng/ml) compared with controls (0.725±0.5108 ng/ml) (P<0.01). Furthermore, MIF concentrations were positively correlated with disease severity in both conditions. The authors concluded that MIF may play a significant role in the pathogenesis of AA and vitiligo, and that targeting MIF could represent a promising therapeutic approach.
In 2020, Oh et al (160) investigated the application of conditioned medium (CM) from human umbilical cord blood-derived mesenchymal stem cells (MSCs) to improve hair growth and developed a method to reliably produce this optimized CM. Their results demonstrated that MIF was essential for the effects of the primed MSC-derived CM and identified it as a key modulator of the hair growth-related protein VEGF in dermal papilla cells. These findings strongly suggest that this method could counteract hair loss and serve as a promising agent for hair restoration.
Vitiligo
Just as AA involves the immune targeting of hair follicles, vitiligo is another organ-specific autoimmune disease, marked by depigmentation due to CD8+ T cell-mediated melanocyte destruction. The average lesion is a totally amelanotic, non-textured and white macule, with progressive disfigurement (161-163). Emerging evidence suggests that vitiligo is associated with a weakened antioxidant system, leading to free radical-mediated melanocyte death or deregulated melanogenesis that may trigger autoimmunity (164). MIF concentrations vary throughout the disease stages, potentially acting as an inflammatory setpoint by regulating the release of proinflammatory molecules (161,162,164).
A Chinese study found that the MIF-173G/C polymorphism and increased serum MIF levels were associated with active non-segmental vitiligo (NSV), which accounts for 90% of vitiligo cases (165). In western Mexico, the-794 CATT 5-8 and -173 G>C MIF polymorphisms are similarly associated with increased NSV risk. Moreover, both serum and in situ MIF levels are correlated with active disease status (161). Research has proposed that MIF may play a fundamental role in skin development (161). Both soluble CD27 (sCD27) and MIF are considered reliable serum biomarkers for disease progression, although sCD27 may offer greater predictive value (162,166).
Vitiligo is currently treated with topical corticosteroids, immunomodulatory agents, vitamin D analogs, antioxidants, phototherapy, laser therapy and surgery. Narrowband UVB (311-313 nm) remains the standard treatment (162,163). Novel therapies targeting MIF-related pathways include JAK inhibitors, N-acetyl-p-benzoquinone imine, isoxazoline imine conjugates and amino acid-benzaldehyde analog conjugates (162,163). JAK inhibitors are currently in phase II trials for topical use in patients with vitiligo (163).
RA
RA is a chronic, invasive autoimmune disease characterized by synovial inflammation, joint erosion and bone resorption. Although its etiology is not fully understood, macrophages are one of the most abundant cell types in the synovium. Their excessive activation, enhanced anti-apoptotic capacity and increased production of proinflammatory cytokines contribute to RA pathogenesis (167-170). MIF plays a central role in sustaining inflammation in RA by promoting the production of proinflammatory cytokines and tissue-degrading enzymes. MIF stimulates MIF synovial fibroblast proliferation, neutrophil chemotaxis and osteoclast differentiation (168). Notably, a Swedish study reported decreasing MIF levels in patients with RA, contrary to the commonly considered association between MIF and RA severity (171). A 2017 meta-analysis including Asian, Latin American and Caucasian populations showed significantly elevated circulating MIF levels in patients with RA and found associations between MIF-173C/G and -794CATT5-8 polymorphisms and disease susceptibility (167). In southern Mexico, these MIF polymorphisms were related to disease activity but not susceptibility, contrasting with findings from Western Mexico (172). Further research is needed to clarify these population-specific discrepancies, particularly in the cell-type-specific transcriptional regulation of MIF variants.
Additionally, MIF-173C/G and-794CATT5-8 polymorphisms also appear to influence Th17-associated cytokine secretion in peripheral blood mononuclear cells from patients with Cushing's syndrome and RA. Their differential expression suggests a regulatory role of MIF promoter haplotypes in inflammatory profiles (173). More recently, MIF has been implicated in the regulation of Th2 cytokines such as IL-25, IL-31 and IL-33, which may have immunomodulatory roles in RA (174).
The discovery of CD74 autoantibodies in the serum of patients with RA supports the role of CD74 as a T-cell antigen in spondyloarthritis, eliciting Th1 and Th17 responses (175). A study examining MIF receptor expression at different stages of RA emphasized the need for further investigation into sCD74 as a MIF decoy receptor, capable of down regulating receptor signaling. The same study identified CXCR7 as a scavenger receptor that stabilizes CXCR4 under inflammatory conditions by preventing its internalization and degradation (176). Since 2018, new treatment options have emerged, including Z-590, a MIF inhibitor, and isopsoralen (IPRN), both showing promise in controlling RA-associated inflammation (168,170).
Advances in the therapeutic strategies targeting MIF
The MIF/D-DT/CD74 axis has emerged as a promising therapeutic target not only in oncology [as extensively reviewed by Valdez et al (177)] but also in autoimmune, inflammatory and fibrotic diseases. Valdez et al (177) provide a comprehensive overview of current research and clinical trials targeting this axis in cancer, emphasizing both its therapeutic potential and the challenges associated with this approach. Their report discusses multiple therapeutic agents, including imalumab, ibudilast and milatuzumab, each demonstrating varying degrees of clinical success. However, key challenges remain, including limited efficacy and the unclear contributions of MIF and oxidized MIF in tumorigenesis (177).
Therapeutic strategies targeting MIF and its homolog D-DT are gaining traction as innovative approaches to overcoming tumor immune evasion and resistance to immune checkpoint inhibitors (ICIs) (178-180). Both cytokines are implicated in several cancer hallmarks, such as proliferation, immune suppression, angiogenesis and metastasis. Inhibition of MIF using small molecules such as 4-IPP has demonstrated potent anti-tumor effects in melanoma models, including tumor burden reduction, downregulation of programmed death ligand-1 and HIF1α as well as metabolic reprogramming. These changes are accompanied by increased CD8⁺ T cell infiltration and a shift in macrophage polarization toward a proinflammatory M1 phenotype (178,179). Similarly, D-DT inhibition reduces proliferation markers and increases apoptotic markers in melanoma cells, although its role is less well characterized (180).
Notably, combination therapies targeting both MIF and D-DT have produced synergistic effects. Dual blockade has been linked to decreased infiltration of MDSCs, increased recruitment of CX3CR1+ patrolling monocytes and depletion of immunosuppressive TAMs, collectively fostering a more immunogenic TME (181). These benefits are further enhanced when dual cytokine inhibition is combined with programmed death-1 or CTLA-4 checkpoint inhibitors, leading to improved tumor control, prolonged survival and durable antitumor memory in murine models of melanoma and CRC (178,181). Supporting these findings, transcriptomic data from patients with melanoma revealed that lower MIF and D-DT expression (or higher CD74:MIF and CD74:D-DT ratios) are correlated with improved overall survival and increased immune cell infiltration, suggesting both prognostic and predictive value (177). An overview of these therapies is provided in Table VI. Collectively, these insights underscore the MIF/D-DT axis as a critical immunoregulatory hub and viable target for enhancing ICI efficacy and overcoming resistance.
![]() |
Table VICancer-related MIF therapeutic approaches. |
Beyond oncology, a growing body of research explores MIF-targeting strategies in other disease contexts. One such agent is IGU, or T-614, which selectively inhibits MIF tautomerase activity. This inhibition reduces B cell proliferation, cytokine release and TNF-α-mediated inflammation. IGU also modulates immune homeostasis by regulating Th17 and Treg cells, enhancing glucocorticoid efficacy, and serving as a steroid-sparing agent. These properties support its application in diseases such as RA, lupus and MS, fibrotic conditions and acute disorders such as acetaminophen-induced injury (182-184). ART, primarily known for its antimalarial properties, also modulates MIF activity. ART reduces parasitemia and improves hemoglobin levels by counteracting MIF, making it a potential therapy for anemia associated with parasitic infections. Additionally, ART exerts immunosuppressive effects by inhibiting MIF signaling and reducing proinflammatory mediators, highlighting its promise in treating inflammatory and autoimmune disorders (185,186). IPRN, a compound derived from Psoralea corylifolia, has shown therapeutic potential in RA by targeting MIF. IPRN inhibits cytokine production, cellular migration and proangiogenic activity in RA fibroblast-like synoviocytes. In vivo studies using collagen-induced arthritis models demonstrate that IPRN reduces paw thickness, arthritis scores and circulating inflammatory cytokines, reinforcing its candidacy as a novel RA therapy (168).
Advancements in small-molecule inhibitors have further broadened the therapeutic landscape of MIF inhibition. Guo et al (187) identified benzopyran and triazole derivatives as selective MIF inhibitors. Benzopyran compounds block MIF-CD74 binding and reduce ERK phosphorylation, while triazoles exhibit dose-dependent inhibition of MIF-induced proliferation. Chen et al (188) explored the connection between MIF, homologous recombination and ferroptosis, identifying MN123 as a non-competitive tautomerase inhibitor that sensitizes cells to ferroptosis when combined with RSL3. In parallel, Li et al (189) investigated ICBP90, a transcriptional activator of MIF, and its interaction with a MIF promoter microsatellite. Their study showed that the small molecule, 1-carbomethoxy-5-formyl-4,6,8-trihydroxyphenazine (CMFT), preferentially downregulates MIF in macrophages with high-expression genotypes.
Taken together, these studies, complemented by the findings of Valdez et al (180), highlight the broad therapeutic promise of MIF inhibition. While challenges remain in targeting the MIF/D-DT/CD74 axis in oncology, the expanding body of evidence across various pathologies suggests that fine-tuned modulation of MIF activity and expression could yield significant clinical benefits. A summary of MIF-targeted therapies in autoimmune and inflammatory conditions is provided in Table VII. Future research should prioritize dosing optimization, long-term safety and the development of rational combination therapies to fully harness the potential of MIF-targeted interventions across cancer, autoimmunity and inflammatory diseases.
![]() |
Table VIIAutoimmunity-related MIF therapeutic approaches. |
Nevertheless, according to current literature and a review of publicly available reports from both the U.S. Food and Drug Administration and the European Medicines Agency, to the best of our knowledge, there are no approved therapeutic options that specifically target MIF currently in clinical use. However, research and development efforts are ongoing. Notably, a 2023 U.S. patent (no. US11584717B2), assigned to Yale University, describes MIF modulators that are presently undergoing in vitro and in vivo testing. Among these, CMFT has been the subject of early-stage evaluation, as reported by Li et al (189). In addition, several existing compounds, such as IGU, ART and IPRN, have demonstrated MIF-modulating or inhibitory effects in the context of autoimmune diseases. Despite this, these agents are not specific MIF inhibitors and instead exert broader immunomodulatory actions, limiting their precision in targeting MIF-driven pathways
Conclusions
MIF has emerged as a uniquely multifunctional cytokine at the crossroads of immunity, inflammation and tissue homeostasis. Unlike other proinflammatory mediators, MIF influences both innate and adaptive immune responses through its diverse receptor interactions (including CD74, CD44, CXCR2, CXCR4 and CXCR7) and enzymatic activity, allowing it to regulate immune cell recruitment, survival and function across a wide range of physiological and pathological contexts. The dual role of MIF as both a protector in homeostatic repair and a driver of chronic inflammation and tumorigenesis sets it apart as a compelling and complex target for therapeutic intervention.
Notably, polymorphisms in the MIF gene (such as-794 CATT5-8 and -173G>C) not only increase susceptibility to autoimmune and oncological diseases but also modulate disease severity and treatment responsiveness, suggesting the potential of MIF as a personalized biomarker. Therapeutic strategies that inhibit MIF activity, such as small-molecule inhibitors (including ISO-1 and 4-IPP), immunomodulatory agents (including IGU) and proteolysis-targeting chimeras, have demonstrated efficacy in preclinical models of cancer, autoimmune disease and neuroinflammation. Despite this progress, key questions remain. The precise regulation of MIF expression, the long-term systemic effects of its inhibition and the dynamics of its interaction with various receptors in distinct tissue microenvironments are areas ripe for exploration. Additionally, the development of receptor-specific inhibitors may unlock new therapeutic windows with reduced side effects.
MIF research is uniquely positioned to reshape current paradigms in immunotherapy and inflammation-driven pathology. As our understanding of its molecular mechanisms deepens, MIF stands not only as a marker of disease but also as a gateway to precision medicine strategies that can modulate immune responses with unprecedented specificity. The continued integration of genetic, biochemical and clinical insights will be essential in translating MIF-targeted therapies into viable, mass-scale solutions across oncology, autoimmunity and regenerative medicine.
Availability of data and materials
Not applicable.
Authors' contributions
GCM and GAN reviewed the literature and wrote the manuscript. RRP, JFMV and UDM reviewed and critically revised the manuscript. LABH defined and supervised the contents of the text and figures generated and conceived the original idea for this review. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Use of artificial intelligence tools
During the preparation of this work, artificial intelligence tools (ChatGPT, https://chatgpt.com) were used to improve the readability and language of the manuscript or to generate images, and subsequently, the authors revised and edited the content produced by the artificial intelligence tools as necessary, taking full responsibility for the ultimate content of the present manuscript.
Acknowledgments
Not applicable.
Funding
This publication was funded by the Program to Support the Improvement of Production Conditions for Members of the SNII and SNCA (grant no. PROSNII 2024-LABH).
References
Bloom BR and Bennett B: Mechanism of a reaction in vitro associated with delayed-type hypersensitivity. Science. 153:80–82. 1966. View Article : Google Scholar : PubMed/NCBI |
|
David JR: Delayed hypersensitivity in vitro: Its mediation by cell-free substances formed by lymphoid cell-antigen interaction. Proc Natl Acad Sci USA. 56:72–77. 1966. View Article : Google Scholar : PubMed/NCBI |
|
Weiser WY, Temple PA, Witek-Giannotti JS, Remold HG, Clark SC and David JR: Molecular cloning of a cDNA encoding a human macrophage migration inhibitory factor. Proc Natl Acad Sci USA. 86:7522–7526. 1989. View Article : Google Scholar : PubMed/NCBI |
|
Bernhagen J, Mitchell RA, Calandra T, Voelter W, Cerami A and Bucala R: Purification, bioactivity, and secondary structure analysis of mouse and human macrophage migration inhibitory factor (MIF). Biochemistry. 33:14144–14155. 1994. View Article : Google Scholar : PubMed/NCBI |
|
Nishino T, Bernhagen J, Shiiki H, Calandra T, Dohi K and Bucala R: Localization of macrophage migration inhibitory factor (MIF) to secretory granules within the corticotrophic and thyrotrophic cells of the pituitary gland. Mol Med. 1:781–788. 1995. View Article : Google Scholar : PubMed/NCBI |
|
Sun HW, Bernhagentt J, Bucalat R and Lolis E: Crystal structure at 2.6-A resolution of human macrophage migration inhibitory factor. Proc Natl Acad Sci USA. 93:5191–5196. 1996. View Article : Google Scholar : PubMed/NCBI |
|
Bacher M, Metz CN, Calandra T, Mayer K, Chesney J, Lohoff M, Gemsa D, Donnelly T and Bucala R: An essential regulatory role for macrophage migration inhibitory factor in T-cell activation. Proc Natl Acad Sci USA. 93:7849–7854. 1996. View Article : Google Scholar : PubMed/NCBI |
|
Chesney J, Metz C, Bacher M, Peng T, Meinhardt A and Bucala R: An essential role for macrophage migration inhibitory factor (MIF) in angiogenesis and the growth of a murine lymphoma. Mol Med. 5:181–191. 1999. View Article : Google Scholar : PubMed/NCBI |
|
Huang XR, Chun Hui CW, Chen YX, Wong BC, Fung PC, Metz C, Cho CH, Hui WM, Bucala R, Lam SK and Lan HY: Macrophage migration inhibitory factor is an important mediator in the pathogenesis of gastric inflammation in rats. Gastroenterology. 121:619–630. 2001. View Article : Google Scholar : PubMed/NCBI |
|
Kleemann R, Kapurniotu A, Frank RW, Gessner A, Mischke R, Flieger O, Jüttner S, Brunner H and Bernhagen J: Disulfide analysis reveals a role for macrophage migration inhibitory factor (MIF) as thiol-protein oxidoreductase. J Mol Biol. 280:85–102. 1998. View Article : Google Scholar : PubMed/NCBI |
|
Matsunaga J, Sinha D, Pannell L, Santist C, Solano F, Wistow GJ and Hearing VJ: Enzyme activity of macrophage migration inhibitory factor toward oxidized catecholamines. J Biol Chem. 274:3268–3271. 1999. View Article : Google Scholar : PubMed/NCBI |
|
Leech M, Metz C, Santos L, Peng T, Holdsworth SR, Bucala R and Morand EF: Involvement of macrophage migration inhibitory factor in the evolution of rat adjuvant arthritis. Arthritis Rheum. 41:910–917. 1998. View Article : Google Scholar : PubMed/NCBI |
|
Leech M, Metz C, Hall P, Hutchinson P, Gianis K, Smith M, Weedon H, Holdsworth SR, Bucala R and Morand EF: Macrophage migration inhibitory factor in rheumatoid arthritis: Evidence of proinflammatory function and regulation by glucocorticoids. Arthritis Rheum. 42:1601–1608. 1999. View Article : Google Scholar : PubMed/NCBI |
|
Petrovsky N, Socha L, Silva D, Grossman AB, Metz C and Bucala R: Macrophage migration inhibitory factor exhibits a pronounced circadian rhythm relevant to its role as a glucocorticoid counter-regulator. Immunol Cell Biol. 81:137–143. 2003. View Article : Google Scholar : PubMed/NCBI |
|
Leng L, Metz CN, Fang Y, Xu J, Donnelly S, Baugh J, Delohery T, Chen Y, Mitchell RA and Bucala R: MIF signal transduction initiated by binding to CD74. J Exp Med. 197:1467–1476. 2003. View Article : Google Scholar : PubMed/NCBI |
|
Chen LC, Lei HY, Liu CC, Shiesh SC, Chen SH, Liu HS, Lin YS, Wang ST, Shyu HW and Yeh TM: Correlation of serum levels of macrophage migration inhibitory factor with disease severity and clinical outcome in dengue patients. Am J Trop Med Hyg. 74:142–147. 2006. View Article : Google Scholar : PubMed/NCBI |
|
Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, et al: MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 13:587–596. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Chuang YC, Su WH, Lei HY, Lin YS, Liu HS, Chang CP and Yeh TM: Macrophage migration inhibitory factor induces autophagy via reactive oxygen species generation. PLoS One. 7:e376132012. View Article : Google Scholar : PubMed/NCBI |
|
Wang Y, Chen Y, Wang C, Yang M, Wang Y, Bao L, Wang JE, Kim B, Chan KY, Xu W, et al: MIF is a 3′ flap nuclease that facilitates DNA replication and promotes tumor growth. Nat Commun. 12:29542021. View Article : Google Scholar |
|
Michelet C: Evolutionary and functional analysis of macrophage migration inhibitory factors in eukaryotes. unpublished PhD thesis. COMUE Université Côte d'Azur; 2020, In French. |
|
Jankauskas SS, Wong DWL, Bucala R, Djudjaj S and Boor P: Evolving complexity of MIF signaling. Cell Signal. 57:76–88. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Sumaiya K, Langford D, Natarajaseenivasan K and Shanmughapriya S: Macrophage migration inhibitory factor (MIF): A multifaceted cytokine regulated by genetic and physiological strategies. Pharmacol Ther. 233:1080242022. View Article : Google Scholar |
|
Merk M, Baugh J, Zierow S, Leng L, Pal U, Lee SJ, Ebert AD, Mizue Y, Trent JO, Mitchell R, et al: The Golgi-associated protein p115 mediates the secretion of macrophage migration inhibitory factor. J Immunol. 182:6896–6906. 2009. View Article : Google Scholar : PubMed/NCBI |
|
Cheng Q, McKeown SJ, Santos L, Santiago FS, Khachigian LM, Morand EF and Hickey MJ: Macrophage migration inhibitory factor increases leukocyte-endothelial interactions in human endothelial cells via promotion of expression of adhesion molecules. J Immunol. 185:1238–1247. 2010. View Article : Google Scholar : PubMed/NCBI |
|
He XX, Yang J, Ding YW, Liu W, Shen QY and Xia HHX: Increased epithelial and serum expression of macrophage migration inhibitory factor (MIF) in gastric cancer: Potential role of MIF in gastric carcinogenesis. Gut. 55:797–802. 2006. View Article : Google Scholar : PubMed/NCBI |
|
Hu CT, Guo LL, Feng N, Zhang L, Zhou N, Ma LL, Shen L, Tong GH, Yan QW, Zhu SJ, et al: MIF, secreted by human hepatic sinusoidal endothelial cells, promotes chemotaxis and outgrowth of colorectal cancer in liver prometastasis. Oncotarget. 6:22410–22423. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Nishihira J, Koyama Y and Mizue Y: Identification of macrophage migration inhibitory factor (MIF) in human vascular endothelial cells and its induction by lipopolysaccharide. Cytokine. 10:199–205. 1998. View Article : Google Scholar : PubMed/NCBI |
|
Pellowe AS, Sauler M, Hou Y, Merola J, Liu R, Calderon B, Lauridsen HM, Harris MR, Leng L and Zhang Y: Endothelial cell-secreted MIF reduces pericyte contractility and enhances neutrophil extravasation. FASEB J. 33:2171–2186. 2019. View Article : Google Scholar : |
|
Farag AGA, Hammam MA, Habib MS, Elnaidany NF and Kamh ME: Macrophage migration inhibitory factor as an incriminating agent in vitiligo. An Bras Dermatol. 93:191–196. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Waeber G, Calandra T, Roduit R, Haefliger JA, Bonny C, Thompson N, Thorens B, Temler E, Meinhardt A, Bacher M, et al: Insulin secretion is regulated by the glucose-dependent production of islet beta cell macrophage migration inhibitory factor. Proc Natl Acad Sci USA. 94:4782–4787. 1997. View Article : Google Scholar : PubMed/NCBI |
|
Guda MR, Rashid MA, Asuthkar S, Jalasutram A, Caniglia JL, Tsung AJ and Velpula KK: Pleiotropic role of macrophage migration inhibitory factor in cancer. Am J Cancer Res. 9:2760–2773. 2019. |
|
Kang I and Bucala R: The immunobiology of MIF: Function, genetics and prospects for precision medicine. Nat Rev Rheumatol. 15:427–437. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Nobre CCG, de Araújo JMG, de Medeiros Fernandes TAA, Cobucci RN, Lanza DCF, Andrade VS and Fernandes JV: Macrophage migration inhibitory factor (MIF): Biological activities and relation with cancer. Pathol Oncol Res. 23:235–244. 2017. View Article : Google Scholar |
|
Osipyan A, Chen D and Dekker FJ: Epigenetic regulation in macrophage migration inhibitory factor (MIF)-mediated signaling in cancer and inflammation. Drug Discov Today. 26:1728–1734. 2021. View Article : Google Scholar : PubMed/NCBI |
|
Zhou Y, Chen H, Liu L, Yu X, Sukhova GK, Yang M, Zhang L, Kyttaris VC, Tsokos GC, Stillman IE, et al: CD74 deficiency mitigates systemic lupus erythematosus-like autoimmunity and pathological findings in mice. J Immunol. 198:2568–2577. 2017. View Article : Google Scholar : PubMed/NCBI |
|
Younan DNA, Agamia N, Elshafei A and Ebeid N: Serum level of macrophage migration inhibitory factor (MIF) in Egyptians with alopecia areata and its relation to the clinical severity of the disease. J Clin Lab Anal. 29:74–79. 2015. View Article : Google Scholar |
|
Llamas-Covarrubias MA, Valle Y, Navarro-Hernández RE, Guzmán-Guzmán IP, Ramírez-Dueñas MG, Rangel-Villalobos H, Estrada-Chávez C and Muñoz-Valle JF: Serum levels of macrophage migration inhibitory factor are associated with rheumatoid arthritis course. Rheumatol Int. 32:2307–2311. 2012. View Article : Google Scholar |
|
Mizue Y, Nishihira J, Miyazaki T, Fujiwara S, Chida M, Nakamura K, Kikuchi K and Mukai M: Quantitation of macrophage migration inhibitory factor (MIF) using the one-step sandwich enzyme immunosorbent assay: Elevated serum MIF concentrations in patients with autoimmune diseases and identification of MIF in erythrocytes. Int J Mol Med. 5:397–403. 2000.PubMed/NCBI |
|
Willeke P, Gaubitz M, Schotte H, Maaser C, Domschke W, Schlüter B and Becker H: Increased serum levels of macrophage migration inhibitory factor in patients with primary Sjögren's syndrome. Arthritis Res Ther. 9:R432007. View Article : Google Scholar |
|
Ren Y, Law S, Huang X, Lee PY, Bacher M, Srivastava G and Wong J: Macrophage migration inhibitory factor stimulates angiogenic factor expression and correlates with differentiation and lymph node status in patients with esophageal squamous cell carcinoma. Ann Surg. 242:55–63. 2005. View Article : Google Scholar : PubMed/NCBI |
|
Fersching DMI, Nagel D, Siegele B, Salat C, Heinemann V, Holdenrieder S and Stoetzer OJ: Apoptosis-related biomarkers sFAS, MIF, ICAM-1 and PAI-1 in serum of breast cancer patients undergoing neoadjuvant chemotherapy. Anticancer Res. 32:2047–2058. 2012.PubMed/NCBI |
|
Yamada G, Shijubo N, Takagi-Takahashi Y, Nishihira J, Mizue Y, Kikuchi K and Abe S: Elevated levels of serum macrophage migration inhibitory factor in patients with pulmonary tuberculosis. Clin Immunol. 104:123–127. 2002. View Article : Google Scholar : PubMed/NCBI |
|
Ganganna A, Subappa A and Bhandari P: Serum migration inhibitory factor levels in periodontal health and disease, its correlation with clinical parameters. Indian J Dent Res. 31:840–845. 2020. View Article : Google Scholar |
|
Emonts M, Sweep FCGJ, Grebenchtchikov N, Geurts-Moespot A, Knaup M, Chanson AL, Erard V, Renner P, Hermans PW, Hazelzet JA and Calandra T: Association between high levels of blood macrophage migration inhibitory factor, inappropriate adrenal response, and early death in patients with severe sepsis. Clin Infect Dis. 44:1321–1328. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Honda K, Nishihira J, Nitta K, Kobayashi H, Uchida K, Kawashima A, Yumura W and Nihei H: Serum levels of macrophage migration inhibitory factor in various types of glomerulonephritis. Nephron. 86:91–92. 2000. View Article : Google Scholar : PubMed/NCBI |
|
Lee TJ, Chun JK, Yeon SI, Shin JS and Kim DS: Increased serum levels of macrophage migration inhibitory factor in patients with Kawasaki disease. Scand J Rheumatol. 36:222–225. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Sugimoto H, Taniguchi M, Nakagawa A, Tanaka I, Suzuki M and Nishihira J: Crystal structure of human D-dopachrome tautomerase, a homologue of macrophage migration inhibitory factor, at 1.54 A resolution. Biochemistry. 38:3268–3279. 1999. View Article : Google Scholar : PubMed/NCBI |
|
Cooke G, Armstrong ME and Donnelly SC: Macrophage migration inhibitory factor (MIF), enzymatic activity and the inflammatory response. Biofactors. 35:165–168. 2009. View Article : Google Scholar : PubMed/NCBI |
|
Fernández G, Arráiz De Fernández C, Valero N, Andrés Martínez J and López J: Adiponectin, hepatic steatosis and the prevention of child obesity. Enferm Inv. 5:17–24. 2020.In Spanish. |
|
Farr L, Ghosh S and Moonah S: Role of MIF cytokine/CD74 receptor pathway in protecting against injury and promoting repair. Front Immunol. 11:12732020. View Article : Google Scholar : PubMed/NCBI |
|
Lue H, Thiele M, Franz J, Dahl E, Speckgens S, Leng L, Fingerle-Rowson G, Bucala R, Lüscher B and Bernhagen J: Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene. 26:5046–5059. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Matza D, Kerem A, Medvedovsky H, Lantner F and Shachar I: Invariant chain-induced B cell differentiation requires intramembrane proteolytic release of the cytosolic domain. Immunity. 17:549–560. 2002. View Article : Google Scholar : PubMed/NCBI |
|
Schneppenheim J, Dressel R, Hüttl S, Lüllmann-Rauch R, Engelke M, Dittmann K, Wienands J, Eskelinen EL, Hermans-Borgmeyer I, Fluhrer R, et al: The intramembrane protease SPPL2a promotes B cell development and controls endosomal traffic by cleavage of the invariant chain. J Exp Med. 210:41–58. 2013. View Article : Google Scholar : |
|
Bucala R and Shachar I: The integral role of CD74 in antigen presentation, MIF signal transduction, and B cell survival and homeostasis. Mini Rev Med Chem. 14:1132–1138. 2014. View Article : Google Scholar |
|
Lue H, Kapurniotu A, Fingerle-Rowson G, Roger T, Leng L, Thiele M, Calandra T, Bucala R and Bernhagen J: Rapid and transient activation of the ERK MAPK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on JAB1/CSN5 and Src kinase activity. Cell Signal. 18:688–703. 2006. View Article : Google Scholar |
|
Gaber T, Schellmann S, Erekul KB, Fangradt M, Tykwinska K, Hahne M, Maschmeyer P, Wagegg M, Stahn C, Kolar P, et al: Macrophage migration inhibitory factor counterregulates dexamethasone-mediated suppression of hypoxia-inducible factor-1 alpha function and differentially influences human CD4+ T cell proliferation under hypoxia. J Immunol. 186:764–774. 2011. View Article : Google Scholar |
|
Mun SH, Oh D and Lee SK: Macrophage migration inhibitory factor down-regulates the RANKL-RANK signaling pathway by activating Lyn tyrosine kinase in mouse models. Arthritis Rheumatol. 66:2482–2493. 2014. View Article : Google Scholar : PubMed/NCBI |
|
Guo Y, Pan W, Liu S, Shen Z, Xu Y and Hu L: ERK/MAPK signalling pathway and tumorigenesis (Review). Exp Ther Med. 19:1997–2007. 2020.PubMed/NCBI |
|
Bucala R and Bernhagen J: MIF Family Cytokines in Innate Immunity and Homeostasis. Springer; Cham: 2018 |
|
Zhang S, Zhao J, Zhang Y, Zhang Y, Cai F, Wang L and Song W: Upregulation of MIF as a defense mechanism and a biomarker of Alzheimer's disease. Alzheimers Res Ther. 11:542019. View Article : Google Scholar : PubMed/NCBI |
|
Heinrichs D, Berres ML, Coeuru M, Knauel M, Nellen A, Fischer P, Philippeit C, Bucala R, Trautwein C, Wasmuth HE and Bernhagen J: Protective role of macrophage migration inhibitory factor in nonalcoholic steatohepatitis. FASEB J. 28:5136–5147. 2014. View Article : Google Scholar : PubMed/NCBI |
|
Bencová S: Transcriptomic analysis of cutaneous inflammatory biomarkers in a mouse model of small fiber neuropathy. Univerzita Karlova, Farmaceutická fakulta v Hradci Králové; 2018 |
|
Miller EJ, Li J, Leng L, McDonald C, Atsumi T, Bucala R and Young LH: Macrophage migration inhibitory factor stimulates AMP-activated protein kinase in the ischaemic heart. Nature. 451:578–582. 2008. View Article : Google Scholar : PubMed/NCBI |
|
Shachar I: An essential MIF-CD74 signaling axis in kidney tubular regeneration, with prospects for precision medicine and pharmacological augmentation. Am J Physiol Renal Physiol. 313:F1084–F1086. 2017. View Article : Google Scholar : PubMed/NCBI |
|
Stoppe C, Averdunk L, Goetzenich A, Soppert J, Marlier A, Kraemer S, Vieten J, Coburn M, Kowark A, Kim BS, et al: The protective role of macrophage migration inhibitory factor in acute kidney injury after cardiac surgery. Sci Transl Med. 10:eaan48862018. View Article : Google Scholar : PubMed/NCBI |
|
Sneath RJS and Mangham DC: The normal structure and function of CD44 and its role in neoplasia. Mol Pathol. 51:191–200. 1998. View Article : Google Scholar |
|
Weng X, Maxwell-Warburton S, Hasib A, Ma L and Kang L: The membrane receptor CD44: Novel insights into metabolism. Trends Endocrinol Metab. 33:318–332. 2022. View Article : Google Scholar : PubMed/NCBI |
|
Noe JT and Mitchell RA: MIF-dependent control of tumor immunity. Front Immunol. 11:6099482020. View Article : Google Scholar : PubMed/NCBI |
|
Calandra T and Roger T: Macrophage migration inhibitory factor: A regulator of innate immunity. Nat Rev Immunol. 3:791–800. 2003. View Article : Google Scholar : PubMed/NCBI |
|
Das R, Koo MS, Kim BH, Jacob ST, Subbian S, Yao J, Leng L, Levy R, Murchison C, Burman WJ, et al: Macrophage migration inhibitory factor (MIF) is a critical mediator of the innate immune response to Mycobacterium tuberculosis. Proc Natl Acad Sci USA. 110:E2997–E3006. 2013. View Article : Google Scholar : PubMed/NCBI |
|
Ruiz-Rosado Jde D, Olguín JE, Juárez-Avelar I, Saavedra R, Terrazas LI, Robledo-Avila FH, Vazquez-Mendoza A, Fernández J, Satoskar AR, Partida-Sánchez S and Rodriguez-Sosa M: MIF promotes classical activation and conversion of inflammatory Ly6C(high) monocytes into TipDCs during murine toxoplasmosis. Mediators Inflamm. 2016:91017622016.PubMed/NCBI |
|
Koga K, Kenessey A, Powell SR, Sison CP, Miller EJ and Ojamaa K: Macrophage migration inhibitory factor provides cardioprotection during ischemia/reperfusion by reducing oxidative stress. Antioxid Redox Signal. 14:1191–1202. 2011. View Article : Google Scholar |
|
Luedike P, Hendgen-Cotta UB, Sobierajski J, Totzeck M, Reeh M, Dewor M, Lue H, Krisp C, Wolters D, Kelm M, et al: Cardioprotection through S-nitros(yl)ation of macrophage migration inhibitory factor. Circulation. 125:1880–1889. 2012. View Article : Google Scholar : PubMed/NCBI |
|
Rassaf T, Weber C and Bernhagen J: Macrophage migration inhibitory factor in myocardial ischaemia/reperfusion injury. Cardiovasc Res. 102:321–328. 2014. View Article : Google Scholar : PubMed/NCBI |
|
Cherepkova OA, Lyutova EM, Eronina TB and Gurvits BY: Chaperone-like activity of macrophage migration inhibitory factor. Int J Biochem Cell Biol. 38:43–55. 2006. View Article : Google Scholar |
|
Israelson A, Ditsworth D, Sun S, Song S, Liang J, Hruska-Plochan M, McAlonis-Downes M, Abu-Hamad S, Zoltsman G, Shani T, et al: Macrophage migration inhibitory factor as a chaperone inhibiting accumulation of misfolded SOD1. Neuron. 86:218–232. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Leyton-Jaimes MF, Benaim C, Abu-Hamad S, Kahn J, Guetta A, Bucala R and Israelson A: Endogenous macrophage migration inhibitory factor reduces the accumulation and toxicity of misfolded SOD1 in a mouse model of ALS. Proc Natl Acad Sci USA. 113:10198–10203. 2016. View Article : Google Scholar : PubMed/NCBI |
|
Apte RS, Sinha D, Mayhew E, Wistow GJ and Niederkorn JY: Cutting edge: Role of macrophage migration inhibitory factor in inhibiting NK cell activity and preserving immune privilege. J Immunol. 160:5693–5696. 1998. View Article : Google Scholar : PubMed/NCBI |
|
Ito T, Ito N, Saatoff M, Hashizume H, Fukamizu H, Nickoloff BJ, Takigawa M and Paus R: Maintenance of hair follicle immune privilege is linked to prevention of NK cell attack. J Invest Dermatol. 128:1196–1206. 2008. View Article : Google Scholar |
|
Matsuda A, Tagawa Y, Matsuda H and Nishihira A: Expression of macrophage migration inhibitory factor in corneal wound healing in rats. Invest Ophthalmol Vis Sci. 38:1555–1562. 1997.PubMed/NCBI |
|
Nishio Y, Minami A, Kato H, Kaneda K and Nishihira J: Identification of macrophage migration inhibitory factor (MIF) in rat peripheral nerves: Its possible involvement in nerve regeneration. Biochim Biophys Acta. 1453:74–82. 1998. View Article : Google Scholar |
|
Shimizu T, Nishihira J, Mizue Y, Nakamura H, Abe R, Watanabe H, Ohkawara A and Shimizu H: High macrophage migration inhibitory factor (MIF) serum levels associated with extended psoriasis. J Invest Dermatol. 116:989–990. 2001. View Article : Google Scholar : PubMed/NCBI |
|
Benigni F, Atsumi T, Calandra T, Metz C, Echtenacher B, Peng T and Bucala R: The proinflammatory mediator macrophage migration inhibitory factor induces glucose catabolism in muscle. J Clin Invest. 106:1291–1300. 2000. View Article : Google Scholar : PubMed/NCBI |
|
Stojanovic I, Saksida T and Stosic-Grujicic S: Beta cell function: The role of macrophage migration inhibitory factor. Immunol Res. 52:81–88. 2012. View Article : Google Scholar : PubMed/NCBI |
|
Bozza FA, Gomes RN, Japiassú AM, Soares M, Castro-Faria-Neto HC, Bozza PT and Bozza MT: Macrophage migration inhibitory factor levels correlate with fatal outcome in sepsis. Shock. 22:309–313. 2004. View Article : Google Scholar : PubMed/NCBI |
|
Kim HR, Kim KW, Jung HG, Yoon KS, Oh HJ, Cho M La and Lee SH: Macrophage migration inhibitory factor enhances osteoclastogenesis through upregulation of RANKL expression from fibroblast-like synoviocytes in patients with rheumatoid arthritis. Arthritis Res Ther. 13:E432011. View Article : Google Scholar |
|
Sanchez-Zamora Y, Terrazas LI, Vilches-Flores A, Leal E, Juárez I, Whitacre C, Kithcart A, Pruitt J, Sielecki T, Satoskar AR and Rodriguez-Sosa M: Macrophage migration inhibitory factor is a therapeutic target in treatment of non-insulin-dependent diabetes mellitus. FASEB J. 24:2583–2590. 2010. View Article : Google Scholar : PubMed/NCBI |
|
Donnelly SC, Hasleit C, Reid PT, Grant IS, Wallace WA, Metz CN, Bruce LJ and Bucala R: Regulatory role for macrophage migration inhibitory factor in acute respiratory distress syndrome. Nat Med. 3:320–323. 1997. View Article : Google Scholar : PubMed/NCBI |
|
Benedek G, Meza-Romero R, Jordan K, Zhang Y, Nguyen H, Kent G, Li J, Siu E, Frazer J, Piecychna M, et al: MIF and D-DT are potential disease severity modifiers in male MS subjects. Proc Natl Acad Sci USA. 114:E8421–E8429. 2017. View Article : Google Scholar : PubMed/NCBI |
|
Tu Y, Guo R, Li J, Wang S, Leng L, Deng J, Bucala R and Lu L: MiRNA regulation of MIF in SLE and attenuation of murine lupus nephritis with miR-654. Front Immunol. 10:22292019. View Article : Google Scholar : PubMed/NCBI |
|
Bezdek S, Leng L, Busch H, Mousavi S, Rades D, Dahlke M, Zillikens D, Bucala R and Sadik CD: Macrophage migration inhibitory factor (MIF) drives murine psoriasiform dermatitis. Front Immunol. 9:22622018. View Article : Google Scholar : PubMed/NCBI |
|
Rajabi F, Amoli MM, Robati RM, Almasi-nasrabadi M and Jabalameli N: Macrophage migration inhibitory factor polymorphism (rs755622) in alopecia areata: A possible role in disease prevention. Arch Dermatol Res. 311:589–594. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Chuang YC, Chen HR and Yeh TM: Pathogenic roles of macrophage migration inhibitory factor during dengue virus infection. Mediators Inflamm. 2015:5470942015. View Article : Google Scholar : PubMed/NCBI |
|
Mohla S: Tumor microenvironment. J Cell Biochem. 101:801–804. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Wang Y and Dong X: MIF improves immune microenvironment of Lewis lung cancer brain metastases after radiotherapy via reducing M2 macrophages. J Clin Oncol. 40(16 Suppl): e210262022. View Article : Google Scholar |
|
Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI |
|
Balogh KN, Templeton DJ and Cross JV: Macrophage migration inhibitory factor protects cancer cells from immunogenic cell death and impairs anti-tumor immune responses. PLoS One. 13:e01977022018. View Article : Google Scholar : PubMed/NCBI |
|
Faucheux L, Grandclaudon M, Perrot-Dockès M, Sirven P, Berger F, Hamy AS, Fourchotte V, Vincent-Salomon A, Mechta-Grigoriou F, Reyal F, et al: A multivariate Th17 metagene for prognostic stratification in T cell non-inflamed triple negative breast cancer. Oncoimmunology. 8:e16241302019. View Article : Google Scholar : PubMed/NCBI |
|
Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI |
|
Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI |
|
Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI |
|
Hashemi Goradel N, Najafi M, Salehi E, Farhood B and Mortezaee K: Cyclooxygenase-2 in cancer: A review. J Cell Physiol. 234:5683–5699. 2019. View Article : Google Scholar |
|
Jäger B, Klatt D, Plappert L, Golpon H, Lienenklaus S, Barbosa PD, Schambach A and Prasse A: CXCR4/MIF axis amplifies tumor growth and epithelial-mesenchymal interaction in non-small cell lung cancer. Cell Signal. 73:1096722020. View Article : Google Scholar : PubMed/NCBI |
|
Cotzomi-Ortega I, Nieto-Yañez O, Juárez-Avelar I, Rojas-Sanchez G, Montes-Alvarado JB, Reyes-Leyva J, Aguilar-Alonso P, Rodriguez-Sosa M and Maycotte P: Autophagy inhibition in breast cancer cells induces ROS-mediated MIF expression and M1 macrophage polarization. Cell Signal. 86:1100752021. View Article : Google Scholar : PubMed/NCBI |
|
Cheng B, Wang Q, Song Y, Liu Y, Liu Y, Yang S, Li D, Zhang Y and Zhu C: MIF inhibitor, ISO-1, attenuates human pancreatic cancer cell proliferation, migration and invasion in vitro, and suppresses xenograft tumour growth in vivo. Sci Rep. 10:67412020. View Article : Google Scholar : PubMed/NCBI |
|
Vaupel P and Mayer A: Hypoxia in tumors: Pathogenesis-related classification, characterization of hypoxia subtypes, and associated biological and clinical implications. Adv Exp Med Biol. 812:19–24. 2014. View Article : Google Scholar : PubMed/NCBI |
|
Avalos-Navarro G, Muñoz-Valle JF, Daneri-Navarro A, Quintero-Ramos A, Franco-Topete RA, Morán-Mendoza AJ, Oceguera-Villanueva A, Bautista-Herrera LA, Topete-Camacho A and Del Toro-Arreola A: Circulating soluble levels of MIF in women with breast cancer in the molecular subtypes: Relationship with Th17 cytokine profile. Clin Exp Med. 19:385–391. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Klemke L, De Oliveira T, Witt D, Winkler N, Bohnenberger H, Bucala R, Conradi LC and Schulz-Heddergott R: Hsp90-stabilized MIF supports tumor progression via macrophage recruitment and angiogenesis in colorectal cancer. Cell Death Dis. 12:1552021. View Article : Google Scholar : PubMed/NCBI |
|
Cotzomi-Ortega I, Rosas-Cruz A, Ramírez-Ramírez D, Reyes-Leyva J, Rodriguez-Sosa M, Aguilar-Alonso P and Maycotte P: Autophagy inhibition induces the secretion of macrophage migration inhibitory factor (MIF) with autocrine and paracrine effects on the promotion of malignancy in breast cancer. Biology (Basel). 9:202020.PubMed/NCBI |
|
Rajendran G, Woolbright BL, Abbott E, Martin A, Dennis K and Taylor JA: Abstract 5150: MIF-2 in bladder cancer: Potential therapeutic target. Cancer Res. 80(16 Suppl): S51502020. View Article : Google Scholar |
|
Richard V, Kindt N and Saussez S: Macrophage migration inhibitory factor involvement in breast cancer (Review). Int J Oncol. 47:1627–1633. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Schabath MB and Cote ML: Cancer progress and priorities: Lung cancer. Cancer Epidemiol Biomarkers Prev. 28:1563–1579. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Guo Y, Hou J, Luo Y and Wang D: Functional disruption of macrophage migration inhibitory factor (MIF) suppresses proliferation of human H460 lung cancer cells by caspase-dependent apoptosis. Cancer Cell Int. 13:282013. View Article : Google Scholar : PubMed/NCBI |
|
Gámez-Pozo A, Sánchez-Navarro I, Calvo E, Agulló-Ortuño MT, López-Vacas R, Díaz E, Camafeita E, Nistal M, Madero R, Espinosa E, et al: PTRF/cavin-1 and MIF proteins are identified as non-small cell lung cancer biomarkers by label-free proteomics. PLoS One. 7:e337522012. View Article : Google Scholar : PubMed/NCBI |
|
Wang WM and Liu JC: Effect and molecular mechanism of mir-146a on proliferation of lung cancer cells by targeting and regulating MIF gene. Asian Pac J Trop Med. 9:806–811. 2016. View Article : Google Scholar : PubMed/NCBI |
|
Huang WC, Kuo KT, Wang CH, Yeh CT and Wang Y: Cisplatin resistant lung cancer cells promoted M2 polarization of tumor-associated macrophages via the Src/CD155/MIF functional pathway. J Exp Clin Cancer Res. 38:1802019. View Article : Google Scholar : PubMed/NCBI |
|
Mawhinney L, Armstrong ME, O'Reilly C, Bucala R, Leng L, Fingerle-Rowson G, Fayne D, Keane MP, Tynan A, Maher L, et al: Macrophage migration inhibitory factor (MIF) enzymatic activity and lung cancer. Mol Med. 20:729–735. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Liu L, Wang J, Wang Y, Chen L, Peng L, Bin Y, Ding P, Zhang R, Tong F and Dong X: Blocking the MIF-CD74 axis augments radiotherapy efficacy for brain metastasis in NSCLC via synergistically promoting microglia M1 polarization. J Exp Clin Cancer Res. 43:1282024. View Article : Google Scholar : PubMed/NCBI |
|
Xiao Z, Song S, Chen D, van Merkerk R, van der Wouden PE, Cool RH, Quax WJ, Poelarends GJ, Melgert BN and Dekker FJ: Proteolysis targeting chimera (PROTAC) for macrophage migration inhibitory factor (MIF) has anti-proliferative activity in lung cancer cells. Angew Chem Int Ed Engl. 60:17514–17521. 2021. View Article : Google Scholar : PubMed/NCBI |
|
Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K and Muschel R: Macrophage migration inhibitory factor: A key cytokine and therapeutic target in colon cancer. Cytokine Growth Factor Rev. 26:451–461. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Shin HN, Moon HH and Ku JL: Stromal cell-derived factor-1α and macrophage migration-inhibitory factor induce metastatic behavior in CXCR4-expressing colon cancer cells. Int J Mol Med. 30:1537–1543. 2012. View Article : Google Scholar : PubMed/NCBI |
|
Lechien JR, Nassri A, Kindt N, Brown DN, Journe F and Saussez S: Role of macrophage migration inhibitory factor in head and neck cancer and novel therapeutic targets: A systematic review. Head Neck. 39:2573–2584. 2017. View Article : Google Scholar : PubMed/NCBI |
|
Grieb G, Merk M, Bernhagen J and Bucala R: Macrophage migration inhibitory factor (MIF): A promising biomarker. Drug News Perspect. 23:257–264. 2010. View Article : Google Scholar : PubMed/NCBI |
|
Zarrin AA, Bao K, Lupardus P and Vucic D: Kinase inhibition in autoimmunity and inflammation. Nat Rev Drug Discov. 20:39–63. 2021. View Article : Google Scholar |
|
Eaton WW, Rose NR, Kalaydjian A, Pedersen MG and Mortensen PB: Epidemiology of autoimmune diseases in Denmark. J Autoimmun. 29:1–9. 2007. View Article : Google Scholar : PubMed/NCBI |
|
Jacobson DL, Gange SJ, Rose NR and Graham NM: Epidemiology and estimated population burden of selected autoimmune diseases in the United States. Clin Immunol Immunopathol. 84:223–243. 1997. View Article : Google Scholar : PubMed/NCBI |
|
Wang L, Wang FS and Gershwin ME: Human autoimmune diseases: A comprehensive update. J Intern Med. 278:369–395. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Rosenblum MD, Remedios KA and Abbas AK: Mechanisms of human autoimmunity. J Clin Invest. 125:2228–2233. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Günther S, Fagone P, Jalce G, Atanasov AG, Guignabert C and Nicoletti F: Role of MIF and D-DT in immune-inflammatory, autoimmune, and chronic respiratory diseases: From pathogenic factors to therapeutic targets. Drug Discov Today. 24:428–439. 2019. View Article : Google Scholar |
|
Brown EM, Kenny DJ and Xavier RJ: Gut microbiota regulation of T cells during inflammation and autoimmunity. Annu Rev Immunol. 37:599–624. 2019. View Article : Google Scholar : PubMed/NCBI |
|
Connelly KL, Kandane-Rathnayake R, Hoi A, Nikpour M and Morand EF: Association of MIF, but not type I interferon-induced chemokines, with increased disease activity in Asian patients with systemic lupus erythematosus. Sci Rep. 6:299092016. View Article : Google Scholar : PubMed/NCBI |
|
Feng X, Chen W, Xiao L, Gu F, Huang J, Tsao BP and Sun L: Artesunate inhibits type I interferon-induced production of macrophage migration inhibitory factor in patients with systemic lupus erythematosus. Lupus. 26:62–72. 2017. View Article : Google Scholar |
|
Fouda ME, El-shimi OS, Mahgoub MY, Abdelrahman SN and Abdelrahman AM: Evaluation of urinary and serum macrophage migration inhibition factor in a group of systemic lupus erythematosus Egyptian patients. Benha Med J. 39:164–178. 2022. |
|
Vincent FB, Slavin L, Hoi AY, Kitching AR, Mackay F, Harris J, Kandane-Rathnayake R and Morand EF: Analysis of urinary macrophage migration inhibitory factor in systemic lupus erythematosus. Lupus Sci Med. 5:e0002772018. View Article : Google Scholar : PubMed/NCBI |
|
Beltrán-Ramírez A, Muñoz-Valle JF, Gamez-Nava JI, Saldaña-Cruz AM, Gonzalez-Lopez L, Padilla-Ortega A, Arias-García FI, Sánchez-Zuno GA, Nava-Valdivia CA, Ponce-Guarneros JM, et al: Steroid resistance associated with high MIF and P-gp serum levels in SLE patients. Molecules. 27:67412022. View Article : Google Scholar : PubMed/NCBI |
|
Nim HT, Connelly K, Vincent FB, Petitjean F, Hoi A, Koelmeyer R, Boyd SE and Morand EF: Novel methods of incorporating time in longitudinal multivariate analysis reveals hidden associations with disease activity in systemic lupus erythematosus. Front Immunol. 10:16492019. View Article : Google Scholar : PubMed/NCBI |
|
De la Cruz-Mosso U, García-Iglesias T, Bucala R, Estrada-García I, González-López L, Cerpa-Cruz S, Parra-Rojas I, Gámez-Nava JI, Pérez-Guerrero EE and Muñoz-Valle JF: MIF promotes a differential Th1/Th2/Th17 inflammatory response in human primary cell cultures: Predominance of Th17 cytokine profile in PBMC from healthy subjects and increase of IL-6 and TNF-α in PBMC from active SLE patients. Cell Immunol. 324:42–49. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Wallace DJ, Wegener WA, Horne H and Goldenberg DM: CT-01 Phase IB study of IMMU-115 (humanised ANTI-CD74 antibody) targeting antigen presenting cells in patients with systemic lupus erythematosus (SLE). Lupus Sci Med. 3:A37–A38. 2016. |
|
Bautista-Herrera LA, De la Cruz-Mosso U, Román-Fernández IV, Parra-Rojas I, Soñanez-Organis JG, Hernández-Bello J, Morales-Zambrano RA, Villanueva-Quintero GD and Muñoz-Valle JF: A potential inflammatory role of IL-31 in psoriatic arthritis: A correlation with Th17 cytokine profile. Int J Immunopathol Pharmacol. 34:20587384209071862020. View Article : Google Scholar : PubMed/NCBI |
|
Morales-Zambrano R, Bautista-Herrera LA, De La Cruz-Mosso U, Villanueva-Quintero GD, Padilla-Gutiérrez JR, Valle Y, Parra-Rojas I, Rangel-Villalobos H, Gutiérrez-Ureña SR and Muñoz-Valle JF: Macrophage migration inhibitory factor (MIF) promoter polymorphisms (-794 CATT5-8 and -173 G>C): association with MIF and TNFα in psoriatic arthritis. Int J Clin Exp Med. 7:2605–2614. 2014. |
|
Schneeberger EE, Citera G, Rodríguez Gil G, Granel A, Arturi A, Rosemffet GM, Maldonado Cocco JA, Berman A, Spindler A and Morales VH: Clinical and immunogenetic characterization in psoriatic arthritis patients. Clin Rheumatol. 34:1413–1418. 2015. View Article : Google Scholar |
|
Barnas JL and Ritchlin CT: Etiology and pathogenesis of psoriatic arthritis. Rheum Dis Clin North Am. 41:643–663. 2015. View Article : Google Scholar : PubMed/NCBI |
|
Veale DJ and Fearon U: The pathogenesis of psoriatic arthritis. Lancet. 391:2273–2284. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Steinhoff M, Meinhardt A, Steinhoff A, Gemsa D, Bucala R and Bacher M: Evidence for a role of macrophage migration inhibitory factor in psoriatic skin disease. Br J Dermatol. 141:1061–1066. 1999. View Article : Google Scholar : PubMed/NCBI |
|
Chhabra S, Banerjee N, Narang T, Sood S, Bishnoi A, Goel S, Bansal F, Singh S, Priyanka K, Minz RW and Dogra S: Single-nucleotide polymorphism and haplotype analysis of macrophage migration inhibitory factor gene and its correlation with serum macrophage migration inhibitory factor levels in North Indian psoriatic patients with moderate disease severity: A cross-sectional study. Indian J Dermatol Venereol Leprol. 89:247–253. 2023. View Article : Google Scholar |
|
Donn RP, Plant D, Jury F, Richards HL, Worthington J, Ray DW and Griffiths CEM: Macrophage migration inhibitory factor gene polymorphism is associated with psoriasis. J Invest Dermatol. 123:484–487. 2004. View Article : Google Scholar : PubMed/NCBI |
|
Dobson R and Giovannoni G: Multiple sclerosis-a review. Eur J Neurol. 26:27–40. 2019. View Article : Google Scholar |
|
Cavalli E, Mazzon E, Basile MS, Mangano K, Di Marco R, Bramanti P, Nicoletti F, Fagone P and Petralia MC: Upregulated Expression of Macrophage Migration Inhibitory Factor, Its Analogue D-Dopachrome Tautomerase, and the CD44 Receptor in Peripheral CD4 T cells from clinically isolated syndrome patients with rapid conversion to clinical defined multiple sclerosis. Medicina (Kaunas). 55:6672019. View Article : Google Scholar |
|
Wanleenuwat P and Iwanowski P: Role of B cells and antibodies in multiple sclerosis. Mult Scler Relat Disord. 36:1014162019. View Article : Google Scholar : PubMed/NCBI |
|
Guan D, Li Y, Cui Y, Zhao H, Dong N, Wang K, Ren D, Song T, Wang X, Jin S, et al: 5-HMF attenuates inflammation and demyelination in experimental autoimmune encephalomyelitis mice by inhibiting the MIF-CD74 interaction. Acta Biochim Biophys Sin (Shanghai). 55:1222–1233. 2023. View Article : Google Scholar : PubMed/NCBI |
|
Hjæresen S, Sejbaek T, Axelsson M, Mortensen SK, Vinsløv-Jensen H, Pihl-Jensen G, Novakova L, Pedersen CB, Halle B, Poulsen FR, et al: MIF in the cerebrospinal fluid is decreased during relapsing-remitting while increased in secondary progressive multiple sclerosis. J Neurol Sci. 439:1203202022. View Article : Google Scholar : PubMed/NCBI |
|
Rijvers L, Melief MJ, van der Vuurst de Vries RM, Stéphant M, van Langelaar J, Wierenga-Wolf AF, Hogervorst JM, Geurts-Moespot AJ, Sweep FCGJ, Hintzen RQ and van Luijn MM: The macrophage migration inhibitory factor pathway in human B cells is tightly controlled and dysregulated in multiple sclerosis. Eur J Immunol. 48:1861–1871. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Castañeda-Moreno VA, De la Cruz-Mosso U, Torres-Carrillo N, Macías-Islas MA, Padilla-De la Torre O, Mireles-Ramírez MA, González-Pérez O, Ruiz-Sandoval JL, Huerta M, Trujillo X, et al: MIF functional polymorphisms (-794 CATT5-8 and -173 G>C) are associated with MIF serum levels, severity and progression in male multiple sclerosis from western Mexican population. J Neuroimmunol. 320:117–124. 2018. View Article : Google Scholar |
|
Vandenbark AA, Meza-Romero R, Benedek G and Offner H: A novel neurotherapeutic for multiple sclerosis, ischemic injury, methamphetamine addiction, and traumatic brain injury. J Neuroinflammation. 16:142019. View Article : Google Scholar : PubMed/NCBI |
|
Bloom J, Metz C, Nalawade S, Casabar J, Cheng KF, He M, Sherry B, Coleman T, Forsthuber T and Al-Abed Y: Identification of iguratimod as an inhibitor of macrophage migration inhibitory factor (MIF) with steroid-sparing potential. J Biol Chem. 291:26502–26514. 2016. View Article : Google Scholar : PubMed/NCBI |
|
Fox RJ, Coffey CS, Conwit R, Cudkowicz ME, Gleason T, Goodman A, Klawiter EC, Matsuda K, McGovern M, Naismith RT, et al: Phase 2 trial of ibudilast in progressive multiple sclerosis. N Engl J Med. 379:846–855. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Pawlitzki M, Sweeney-Reed CM, Meuth SG, Reinhold D and Neumann J: CSF macrophage migration inhibitory factor levels did not predict steroid treatment response after optic neuritis in patients with multiple sclerosis. PLoS One. 13:e02077262018. View Article : Google Scholar : PubMed/NCBI |
|
Eldesouky F, Ibrahim AM and Sharaf SM: Macrophage migration inhibitory factor in alopecia areata and vitiligo: A case-controlled serological study. J Clin Aesthet Dermatol. 13:24–27. 2020. |
|
Sandoval DA and Plaza NE: Collapse of the immunoprivilege and emotional stress in alopecia areata. Rev Argent Dermatol. 101:41–50. 2020. |
|
Oh HA, Kwak J, Kim BJ, Jin HJ, Park WS, Choi SJ, Oh W and Um S: Migration inhibitory factor in conditioned medium from human umbilical cord blood-derived mesenchymal stromal cells stimulates hair growth. Cells. 9:13442020. View Article : Google Scholar : PubMed/NCBI |
|
Garcia-Orozco A, Martinez-Magaña IA, Riera-Leal A, Muñoz-Valle JF, Martinez-Guzman MA, Quiñones-Venegas R, Sánchez-Zuno GA and Fafutis-Morris M: Macrophage inhibitory factor (MIF) gene polymorphisms are associated with disease susceptibility and with circulating MIF levels in active non-segmental vitiligo in patients from western Mexico. Mol Genet Genomic Med. 8:e14162020. View Article : Google Scholar : PubMed/NCBI |
|
Ibrahim HM, Abd El-Aziz Nada EED, Abdel-Hamid Ali S, Hegazy EM and Hassan MH: Macrophage migration inhibitory factor in vitiligo: Pathogenesis and potential therapeutic aspects. Asian J Biochem Genet Mol Biol. 8:8–24. 2021. View Article : Google Scholar |
|
Solimani F, Meier K and Ghoreschi K: Emerging topical and systemic JAK inhibitors in dermatology. Front Immunol. 10:28472019. View Article : Google Scholar : PubMed/NCBI |
|
Chakraborty A, Thulasamma S, Kumar GS, Pallapolu P, Lahari K, Rafeeqi T, Husain G, Jabeen F, Javed G, Waheed MA and Kazmi MH: Expression levels of MIF, NLRP1 and FOXP3 genes along with biomarker levels in patients with active form of non-segmental generalized vitiligo: A study in South Indian population. Songklanakarin J Sci Technol. 44:353–360. 2022. |
|
Wang D, Min S, Lin X and Jiang G: Association among MIF, IFIH1, and IL6 gene polymorphisms and non-segmental vitiligo in a chinese han population. Clin Cosmet Investig Dermatol. 15:1597–1609. 2022. View Article : Google Scholar : PubMed/NCBI |
|
ElGhareeb MI, El Mokadem S, El Sayed B and Khalifa N: Soluble CD27 and MIF as possible serum biomarkers of vitiligo activity in Egyptian patients in Sharkia governorate. Dermatol Reports. 11:82652019. |
|
Bae SC and Lee YH: Associations between circulating macrophage migration inhibitory factor (MIF) levels and rheumatoid arthritis, and between MIF gene polymorphisms and disease susceptibility: A meta-analysis. Postgrad Med J. 94:109–115. 2018. View Article : Google Scholar |
|
Han Y, Wang J, Li S, Li Y, Zhang Y, Zhang R, Zhang Y, Fan H, Shi H, Pan J, et al: Isopsoralen ameliorates rheumatoid arthritis by targeting MIF. Arthritis Res Ther. 23:2432021. View Article : Google Scholar : PubMed/NCBI |
|
Taghadosi M, Adib M, Jamshidi A, Mahmoudi M and Farhadi E: The p53 status in rheumatoid arthritis with focus on fibroblast-like synoviocytes. Immunol Res. 69:225–238. 2021. View Article : Google Scholar : PubMed/NCBI |
|
Zhang Z, Zhang R, Li L, Zhu L, Gao S, Lu Q, Gu Y, Zhang Y, Yang H, Hou T, et al: Macrophage migration inhibitory factor (MIF) inhibitor, Z-590 suppresses cartilage destruction in adjuvant-induced arthritis via inhibition of macrophage inflammatory activation. Immunopharmacol Immunotoxicol. 40:149–157. 2018. View Article : Google Scholar : PubMed/NCBI |
|
Södergren A, Kar p K, Bengtsson C, Möller B, Rantapää-Dahlqvist S and Wållberg-Jonsson S: Biomarkers associated with cardiovascular disease in patients with early rheumatoid arthritis. PLoS One. 14:e02205312019. View Article : Google Scholar : PubMed/NCBI |
|
Santoscoy-Ascencio G, Baños-Hernández CJ, Navarro-Zarza JE, Hernández-Bello J, Bucala R, López-Quintero A, Valdés-Alvarado E, Parra-Rojas I, Illades-Aguiar B and Muñoz-Valle JF: Macrophage migration inhibitory factor promoter polymorphisms are associated with disease activity in rheumatoid arthritis patients from Southern Mexico. Mol Genet Genomic Med. 8:e10372020. View Article : Google Scholar : |
|
Hernández-Palma LA, García-Arellano S, Bucala R, Llamas-Covarrubias MA, De la Cruz-Mosso U, Oregon-Romero E, Cerpa-Cruz S, Parra-Rojas I, Plascencia-Hernández A and Muñoz-Valle JF: Functional MIF promoter haplotypes modulate Th17-related cytokine expression in peripheral blood mononuclear cells from control subjects and rheumatoid arthritis patients. Cytokine. 115:89–96. 2019. View Article : Google Scholar |
|
García-Arellano S, Hernández-Palma LA, Cerpa-Cruz S, Sánchez-Zuno GA, Herrera-Godina MG and Muñoz-Valle JF: The novel role of MIF in the secretion of IL-25, IL-31, and IL-33 from PBMC of patients with rheumatoid arthritis. Molecules. 26:49682021. View Article : Google Scholar : PubMed/NCBI |
|
Sogkas G, Klose K, Baerlecken N, Schweikhard E, Matthias T, Kniesch K, Schmidt RE and Witte T: CD74 is a T cell antigen in spondyloarthritis. Clin Exp Rheumatol. 38:195–202. 2020. View Article : Google Scholar |
|
Sánchez-Zuno GA, Bucala R, Hernández-Bello J, Román-Fernández IV, García-Chagollán M, Nicoletti F, Matuz-Flores MG, García-Arellano S, Esparza-Michel JA, Cerpa-Cruz S, et al: Canonical (CD74/CD44) and non-canonical (CXCR2, 4 and 7) MIF receptors are differentially expressed in rheumatoid arthritis patients evaluated by DAS28-ESR. J Clin Med. 11:1202021. View Article : Google Scholar |
|
Valdez CN, Sánchez-Zuno GA, Bucala R and Tran TT: Macrophage migration inhibitory factor (MIF) and D-dopachrome tautomerase (DDT): Pathways to tumorigenesis and therapeutic opportunities. Int J Mol Sci. 25:48492024. View Article : Google Scholar : PubMed/NCBI |
|
de Azevedo RA, Shoshan E, Whang S, Markel G, Jaiswal AR, Liu A, Curran MA, Travassos LR and Bar-Eli M: MIF inhibition as a strategy for overcoming resistance to immune checkpoint blockade therapy in melanoma. Oncoimmunology. 9:18469152020. View Article : Google Scholar : PubMed/NCBI |
|
Mora Barthelmess R, Stijlemans B and Van Ginderachter JA: Hallmarks of cancer affected by the MIF cytokine family. Cancers (Basel). 15:3952023. View Article : Google Scholar : PubMed/NCBI |
|
Valdez CN, Athziri Sánchez-Zuno G, Osmani L, Ibrahim W, Galan A, Bacchiocchi A, Halaban R, Kulkarni RP, Kang I, Bucala R and Tran T: Prognostic and therapeutic insights into MIF, DDT, and CD74 in melanoma. Oncotarget. 15:507–520. 2024. View Article : Google Scholar : PubMed/NCBI |
|
Sánchez-Zuno GA, Valdez CN, Osmani L, Kang I, Bucala R and Tran T: 571 Dual inhibition of MIF and DDT enhances the efficacy of anti-PD-1 therapy in murine melanoma. J Immunother Cancer. 12:A6512024. |
|
Bloom J, Pantouris G, He M, Aljabari B, Mishra L, Manjula R, Parkins A, Lolis EJ and Al-Abed Y: Iguratimod, an allosteric inhibitor of macrophage migration inhibitory factor (MIF), prevents mortality and oxidative stress in a murine model of acetaminophen overdose. Mol Med. 30:432024. View Article : Google Scholar : PubMed/NCBI |
|
Jiang H, Gao H, Wang Q, Wang M and Wu B: Molecular mechanisms and clinical application of iguratimod: A review. Biomed Pharmacother. 122:1097042020. View Article : Google Scholar : PubMed/NCBI |
|
Long Z, Zeng L, He Q, Yang K, Xiang W, Ren X, Deng Y and Chen H: Research progress on the clinical application and mechanism of iguratimod in the treatment of autoimmune diseases and rheumatic diseases. Front Immunol. 14:11506612023. View Article : Google Scholar : PubMed/NCBI |
|
Efferth T and Oesch F: The immunosuppressive activity of artemisinin-type drugs towards inflammatory and autoimmune diseases. Med Res Rev. 41:3023–3061. 2021. View Article : Google Scholar : PubMed/NCBI |
|
Ruwizhi N, Maseko RB and Aderibigbe BA: Recent advances in the therapeutic efficacy of artesunate. Pharmaceutics. 14:5042022. View Article : Google Scholar : PubMed/NCBI |
|
Guo S, Zhao Y, Yuan Y, Liao Y, Jiang X, Wang L, Lu W and Shi J: Progress in the development of macrophage migration inhibitory factor small-molecule inhibitors. Eur J Med Chem. 286:1172802025. View Article : Google Scholar : PubMed/NCBI |
|
Chen D, Zhao C, Zhang J, Knol CWJ, Osipyan A, Majerníková N, Chen T, Xiao Z, Adriana J and Griffith AJ: Small molecule MIF modulation enhances ferroptosis by impairing DNA repair mechanisms. Adv Sci (Weinh). 11:e24039632024. View Article : Google Scholar : PubMed/NCBI |
|
Li J, Leng L, Pantouris G, Manjula R, Piecychna M, Abriola L, Hu B, Lolis E, Armstrong ME, Donnelly SC and Bucala R: A small-molecule allele-selective transcriptional inhibitor of the MIF immune susceptibility locus. J Biol Chem. 300:1074432024. View Article : Google Scholar : PubMed/NCBI |
|
Sanchez-Zuno GA, Caulfield J, Leng L, Zhang L, Jilaveanu L, Kluger H, Bucala R and Tran T: 830 Inhibition of macrophage migration inhibitory factor (MIF) to overcome immune checkpoint resistance in melanoma. J Immunother Cancer. 11(Suppl 1): S9292023. |