Abstract
The macrophage-migration-inhibitory-factor (MIF)-family consists of the structurally homologous proteins MIF, D-dopachrome tautomerase (D-DT), and D-DT like (D-DTL). While MIF is the most well-described member, much less is known about D-DT, and very little about D-DTL. Here, we provide an overview of the structure, similarities, and biological functions of these proteins. MIF and D-DT can have both protective and aggravating effects on various diseases depending on the disease type, involved organ, cell type, and disease stage. Given that the pathological consequence of many chronic diseases is fibrosis, we here discuss the role of these proteins in organ fibrosis, particularly of the kidney, liver, heart, lung, and skin. We discuss the various roles of these proteins, suggesting that MIF might have pro- and antifibrotic roles in different organs. To date, D-DT has been shown to have only antifibrotic roles. We tackle potential translational considerations and propose future research avenues to better understand the involvement of MIF-family in organ fibrosis.
Keywords: D-DT, D-DTL, CD74, clinical trials
2. Introduction
Cytokines are small, secreted proteins affecting interactions and communications between cells (1). They are modulators of inflammation that participate in acute and chronic inflammation (2). The macrophage migration inhibitory factor (MIF)-family consists of three cytokines: MIF, D-dopachrome tautomerase (D-DT) and D-dopachrome tautomerase like (D-DTL). MIF was first identified in 1966 (3, 4), followed by D-DT in 1993 (5). The structural homology of D-DT and MIF was revealed in 1999 (6).
MIF and D-DT both bind to the extracellular domain of the HLA class II histocompatibility antigen γ chain (Cluster of differentiation (CD)74) (7, 8). As a receptor, CD74 is localized in the membrane but also exists in soluble form after shedding the CD74 ectodomain (sCD74) (9). It is hypothesized that sCD74 might act as a self-regulatory mechanism, or soluble “ligand trap”, preventing the interaction of MIF and D-DT with the CD74 receptor on the cell surface (10, 11). MIF and D-DT also bind to the CXC motif chemokine receptors (CXCR) 4 and CXCR7, and MIF additionally to CXCR2 (12–16). MIF and D-DT are the only known ligands for CD74, whereas the CXCRs have several other ligands (17). This review will focus on the CD74-mediated effects of MIF/D-DT, given that these effects were more specifically investigated compared to those potentially mediated by the CXCRs.
Both MIF and D-DT play a role in various diseases (18–23). This includes inflammatory and chronic conditions, which often lead to organ fibrosis (24). Organ fibrosis represents a significant challenge in medicine, contributing to 17.8% of all global deaths in 2019 with little available effective and specific treatment options (25, 26). Fibrosis is defined by excessive accumulation of fibrous connective tissue (24). It is a physiologically important repair process in certain situations and might have an evolutionary advantage in survival, e.g., in wound repair (27). If fibrosis occurs as a maladaptive repair after acute or chronic tissue damage, it is considered a pathological process (28). During that process, functional tissue is replaced by non-functional connective extracellular matrix (ECM), e.g. collagen or elastin (29). This leads to permanent scarring and organ dysfunction, eventually leading to organ failure and death (24).
Here, we focus on the roles of the MIF-family in organ fibrosis, including kidney, liver, heart, pulmonary, and systemic sclerosis.
2.1. Properties of the MIF-family members and the CD74 receptor
The cytokines belonging to the MIF-family exhibit a high degree of homology (Figure 1). MIF, D-DT, and D-DTL consist of three exons of almost identical size, and only the non-coding introns have different lengths (30) (Figure 1). At the protein level, the amino acid sequences of D-DT and MIF match 34% and 27% in humans and mice, respectively (30). The most recently reported member, D-DTL, shows a 56% mRNA sequence identity to D-DT and a 21% sequence identity to MIF in humans. Currently, it is not known whether D-DTL is expressed in mice. Human MIF, D-DT, and D-DTL are closely linked on chromosome 22 in humans (31, 32) and MIF and D-DT on chromosome 10 in mice (31) (Figure 1a+f+j)
Figure 1. Overview of the MIF-family.
The overview includes the properties of the proteins macrophage migration inhibitors factor (MIF), D-dopachrome tautomerase (D-DT) and D-DT like (D-DTL) in the category: gene, structure, receptors, and CD74 binding. Due to the limited data available on D-DTL, unknown properties are marked with a question mark. Created in BioRender. Boor, P. (2025) https://BioRender.com/q16n769.
2.1.1. Macrophage-migration-inhibitory-factor (MIF)
MIF is an enzyme having an N-terminal enzymatic site that can tautomerize p-hydroxyphenylpyruvate (33, 34). The tautomerase function is not essential for its biological role and is probably a rudimentary property of MIF that derives from its original function in invertebrate immunity (30, 35). However, the catalytic base proline (Pro-1) also plays a structural role in receptor binding, making it a relevant target for the development of small-molecule MIF inhibitors (36).
MIF was named after the initial observation to inhibit macrophage migration during allergic reactions (3, 19). Despite its early discovery, most studies show an effect in support of directed migration and recruitment of leukocytes (19, 37). Like chemokines, MIF induces integrin-dependent arrest and transmigration of monocytes and T cells (38). Important for the chemokine-like function is its pseudo(E)LR domain (39). Under physiological conditions, three MIF monomers build a homotrimer (40, 41) (Figure 1c). As a homotrimer, MIF binds to its receptors (42, 43) (Figure 1c).
MIF binds with a high affinity to the extracellular domain of CD74 (7). MIF homotrimer is kept in place by three CD74 molecules, resulting in a long-lasting complex with a slow association and a slow dissociation constant (43) (Figure 1d). In addition, MIF binds to the non-cognate CXCR2, CXCR4, and CXCR7 (13, 44) (Figure 1c). This binding does not appear specific to MIF, as other cytokines can also bind to CXCRs (15). It modulates the migration of immune cells through the CXCRs (30). MIF was shown to directly activate extracellular signal-regulated kinase (ERK)/ protein kinase B (AKT) or transforming growth factor-β (TGF-β)/SMADs signaling and indirectly promote the production or expression of a large panel of proinflammatory molecules, including cytokines (Tumor necrosis factor (TNF), interferon-γ, interleukin (IL)-1β, IL-2, IL-6, IL-8), nitric oxide, COX2, and several matrix metalloproteinases and their inhibitors (45–48). It also acts as a key upstream regulator in the inflammatory cascade, and toll-like receptor-4-mediated pathways were shown to be involved in the regulation of MIF expression (48). MIF’s synergy with damage-associated molecular patterns was not explicitly demonstrated with respect to fibrosis.
MIF exists in two immunologically distinct conformational isoforms, reduced (redMIF) and oxidized (oxMIF). RedMIF is present in plasma, cells, and tissue of healthy and diseased patients. It converts into oxMIF in an oxidized, inflammatory environment (49, 50). OxMIF was found in the plasma and tissue of patients with inflammatory diseases and in solid tumors. Therapeutic antibodies have been shown to selectively bind oxMIF but not redMIF. (49) The differentiation between oxMIF and redMIF plays a crucial role in therapy, which we discuss in more detail in the chapter “Interventional clinical trials” (Chapter 2.3.). In the subsequently described studies of MIF in organ fibrosis (Chapter 2.2.), a differentiation between oxMIF and redMIF is not made, as no study distinguishes between both.
2.1.2. D-Dopachrome tautomerase (D-DT)
D-DT shares structural similarities with MIF; therefore, it is also named MIF-2 (6, 51). It has an N-terminal enzymatic site that can tautomerize D-dopachrome to 5,6-dihydroxyindole (52). The tautomerase activity of D-DT is considered a potential evolutionary relic and, like MIF, the catalytic base proline (Pro-1) plays an important role in receptor binding (30).
D-DT accumulates in active form as a homotrimer (43) (Figure 1g+h). The C-terminal region is key in molecular recognition because it can regulate the active site opening, protein-ligand interactions, and conformational flexibility of the catalytic pocket’s environment (53).
D-DT binds to CD74, but with lower affinity compared to MIF (8). Topologically, the location of the three CD74 binding regions of the D-DT homotrimer differs substantially from that of the three MIF binding regions (43) (Figure 1i). This key difference in orientation appears to derive from an eight amino acid sequence insertion in D-DT, that topologically limits the binding of D-DT homotrimer to only one CD74 (43) (Figure 1i), resulting in a weaker interaction with a rapid complex formation but also a rapid dissociation rate (43). Molecular simulations reveal that D-DT contains a hydrogen bond network comparable to that of MIF (51).
D-DT also binds to CXCR4 (23) and CXCR7 (16) (Figure 1h). However, D-DT does not bind to CXCR2 because it lacks the pseudo(E)LR (Arg11, Asp4) domain of MIF (54). In the heart, it has been shown that the lack of interaction with CXCR2 abolishes the negative inotropic effect of MIF, which reduces cardiac contractility (12). The missing domain leads to decreased recruitment of monocytes and leukocytes in contrast to MIF (12).
2.1.3. D-Dopachrome tautomerase like (D-DTL)
D-DTL is predicted to enable phenylpyruvate tautomerase activity and be involved in melanin biosynthetic processes. However, it is not clear if the gene is expressed to yield D-DTL protein in humans, and the biological function of this putative protein is unknown (32) (Figure 1).
2.1.4. Cluster of differentiation 74 (CD74)
CD74, the high-affinity receptor of MIF and D-DT, forms a trimer (7, 8). Three CD74 trimers are bound to three binding sites of MIF, and one CD74 trimer is bound to one binding site of D-DT (43). CD74 lacks an intracellular signaling domain (55). Therefore, a co-receptor, CD44, is required, which provides the necessary intracytoplasmic signaling domain (55). By recruiting the co-receptor CD44, the MIF-family initiates the pathway to stimulate cellular effects (30). For example, stimulation of the membrane-bound CD74, together with CD44, activates the ERK1/2/mitogen-activated protein kinase signaling pathway, the phosphatidylinositol 3-kinase/ AKT signal transduction cascade, nuclear factor-κB (NF-κB), or the AMP-activated protein kinase (AMPK) signaling pathway (56, 57). CD74 is required for MIF-induced activation of the ERK1/2/MAP kinase cascade and cell proliferation (58). The ERK pathway, in general, leads to cell proliferation, including fibroblasts, which could have direct effects on fibrosis (59). Currently, the role of MIF-induced ERK activation in fibrosis is not well known.
CD74 is not only found at the cell membrane, but it also exists in a shed, soluble form of the CD74 ectodomain (sCD74) by regulated intramembrane proteolysis (9, 60) (Figure 1c+h). CD74 undergoes several proteolytic events in the endocytic compartments, removing the bulk of the luminal domain and forming a membrane-bound 1–82 truncated protein (60). The biological role of sCD74 is not fully understood, but it is hypothesized that sCD74 acts as a self-regulatory mechanism that prevents the interaction of MIF and D-DT with the CD74 receptor on the cell surface, i.e., functioning as an endogenous “ligand-trap” known for some other receptors (10, 11). D-DT reduced MIF binding to the sCD74 ectodomain in a dose-dependent manner (39). It exhibits a 3-fold higher association rate to sCD74 but an 11-fold faster dissociation rate than MIF (39). In contrast to MIF, D-DT shows a high flexibility of the C-terminus, which can be triggered, for example, by phosphorylation (61).
2.1.5. Expression of the MIF family
Here, we discuss the expression of MIF, D-DT, and D-DTL in various organs (Figure 2) and individual cell types (Figure 3).
Figure 2. RNA and protein expression comparison of the MIF-family, and CD74.
(A) In a heat map, the RNA expression of MIF, D-DT, D-DTL, and CD74 was screened in different tissues. The RNA-seq tissue data generated as part of the Genotype-Tissue Expression (GTEx) project are indicated as nTPM (normalized protein-coding transcripts per million), corresponding to the mean values of the various individual samples from each tissue. The nTPM values got normalized for each organ. (B) MIF, D-DT, and CD74 protein expression were compared in immunohistochemistry staining of the kidney, liver, heart, lung, and skin. D-DTL was excluded because of insufficient staining. Scalebar: 50 μm. The RNA and protein data are sourced from the Human Protein Atlas (72). Image/gene/data available from v24.0.proteinatlas.org. Created in BioRender. Boor, P. (2025) https://BioRender.com/w32f754.
Figure 3. Expression of the MIF-family and CD74 of the respective cell types in different tissues.
The data originates from single-cell RNA analyses. RNA expression of each cluster is indicated as nTPM (normalized protein-coding transcripts per million) from each cell type from the kidney, liver, heart, lung, and skin. The single-cell RNA sequencing data are sourced from the Human Protein Atlas (72). Data are available from v24.0.proteinatlas.org. The cluster data was summarized by taking the mean value of the same cell types. Created in BioRender. Boor, P. (2025) https://BioRender.com/3h6zrcy.
2.1.5.1. Expression of MIF
MIF is located intracellularly, and it gets secreted (62). MIF lacks a signal sequence and is secreted by an unconventional secretion mediated by Golgi-associated protein p115 (63). MIF RNA is strongly expressed by a broad spectrum of organs (Figure 2A). The highest tissue-specific RNA expression of MIF is found in the kidney (Figure 2A).
Under homeostatic conditions, MIF is constitutively and ubiquitously expressed (64). Stimulation of macrophages with lipopolysaccharide (LPS) leads in vitro to the release of MIF, as the in vivo stimulation of mice resulted in a time-dependent increase of MIF (39). It is expressed by immune cells, but also, for example, by fibroblasts (65), endothelial (66), neuroendocrine (67), mesenchymal cells (68), or epithelial cells (69) (Figure 3).
Some data suggest sexual dimorphism of MIF. Neonatal immune signatures displayed higher levels of MIF in newborn male blood spots compared to females (70). In a clinical trial, MIF was positively correlated with testosterone and negatively with estradiol (71). Higher MIF levels are identified in male multiple sclerosis patients compared to female multiple sclerosis patients (20). Suggesting a sex difference between males and females. Despite these few studies, the sexual dimorphism of MIF represents an important gap in knowledge.
2.1.5.2. Expression of D-DT
The mRNA of D-DT lacks an N-terminal or an internal secretory signal sequence, suggesting that D-DT is secreted via a specialized, non-classical export pathway (39). D-DT is located intracellularly (39). D-DT’s highest tissue-specific RNA expression is detected in the liver (Figure 2A). D-DT RNA is also highly expressed in the pancreas and kidney, while expression in the remaining organs is moderate to low (Figure 2A).
Macrophages stimulated with LPS secrete 20-fold more MIF than D-DT (39). Furthermore, MIF shows a steeper dose response in the measurement of macrophage migration inhibition and glucocorticoid overdrive than D-DT (30). Like MIF, D-DT is expressed not only by immune cells but also by epithelial cells (16, 22). (Figure 3)
Sexual dimorphisms were also detected for D-DT. Similarly to MIF, higher D-DT levels are identified in male multiple sclerosis patients compared to female multiple sclerosis patients (20).
2.1.5.3. Expression of D-DTL
D-DTL was predicted to have an intracellular localization (72). Very little, if any D-DTL RNA is found in various organs (Figure 2A), with the highest expression in the liver (Figure 2A + Figure 3). There is no data available on sexual dimorphism for D-DTL.
2.1.5.4. Expression of CD74
The strongest CD74 expression is observed in cells involved in the antigen-presenting cells, like dendritic cells, B cells, and macrophages (73). CD74 functions as a chaperone for the correct folding of major histocompatibility complex class II and regulates its antigen-binding capacity (74). Non-antigen-presenting cells, such as epithelial cells, express CD74 on their surface in various organs, including the kidney, lung, intestine, heart, liver, and skin (56). The strongest RNA expression is detected in the spleen and lungs (Figure 2A). The function of CD74 in epithelial cells remains incompletely elucidated.
In late-stage pancreatic ductal adenocarcinoma, a unique fibroblast population expressing the major histocompatibility complex class II genes was identified (75). CD74-expressing antigen-presenting fibroblasts have also been identified in multiple cancer types, with diverse roles ranging from promoting immunosuppression to enhancing anti-tumor immunity, depending on the specific cancer type (76).”
2.2. The pro- and antifibrotic effect of the MIF-family in organ fibrosis
CD74 is involved in MIF-family-driven immune cell recruitment and activation of a variety of cellular responses, including cell proliferation (77). The transformation of quiescent fibroblasts into actively proliferating, ECM-producing myofibroblasts plays a pivotal role in the pathogenesis of all fibrotic diseases (78). MIF is known to increase and promote fibroblast proliferation during chronic inflammatory diseases (79). However, MIF also exerts an antifibrotic effect, which demonstrates the pleiotropic effect of MIF (80). D-DT also has pleiotropic functions, including a multifaceted role in organ fibrosis. Here we discuss the relevance of MIF and D-DT in kidney, heart, pulmonary, liver fibrosis, and systemic sclerosis (Table 1). D-DTL is not considered since nothing is known to date.
Table 1. Summary of the expression and functional effects of MIF and D-DT in organ fibrosis.
| Kidney | Liver | Heart | Lung | Skin | ||
|---|---|---|---|---|---|---|
| Circulating level | MIF | CKD/SLE patients: MIF increased | Decompensated cirrhosis: MIF increased, low sCD74 Autoimmune hepatitis/primary biliary cirrhosis: elevated MIF expression | ? | IPF: Serum MIF concentration was associated with a higher 3-month mortality CF: Carrying at least one 5-CATT repeat allele exhibit low MIF expression | SSc: Increased MIF / no difference dSSc: Increased MIF |
| D-DT | ? | ? | ? | ? | SSc: No difference SSc with FEV-1: D-DT increased | |
| Tissue Expression | MIF | Healthy: MIF mainly in tubular epithelial cells Tubulointerstitial fibrosis: MIF decreased | Healthy: MIF Hepatocytes, hepatic stellate cells Autoimmune hepatitis/primary biliary cirrhosis: elevated MIF expression | Healthy: MIF low levels cardiomyocytes Acute myocardial infarction: MIF increased Ischemia: MIF increased Atrial fibrillation: MIF increased | Healthy: MIF mainly in bronchiolar and alveolar epithelium IPF: increased MIF | SSc: MIF increased in keratinocytes and dermal fibroblasts, decreased in endothelial cells dSSc: MIF and CD74 increased om dermal fibroblasts |
| D-DT | Healthy: D-DT mainly in tubular epithelial cells Tubulointerstitial fibrosis: D-DT decreased | ? | Failing hearts: Decreased D-DT TAC: Increased D-DT | ? | ? | |
| Functional role: Investigated models | MIF | UUO, I/R, in vitro | TAA, CCl4, hepatitis B virus-related liver fibrosis, in vitro | Ang-II, in vitro | Bleomycin, in vitro | In vitro |
| D-DT | UUO, I/R | CCl4 | TAC | ? | ? | |
| Functional role: Expression in animal models | MIF | UUO or I/R: MIF decreased | Hepatitis B virus- related liver fibrosis: MIF expression correlated with the activation of fibrosis marker | ? | Bleomycin: increased MIF in bronchoalveolar lavage and fibrotic area | ? |
| D-DT | UUO or I/R: D-DT decreased | CCl4: D-DT increased | ? | ? | ? | |
| Functional role: Neutralization/ Downregulation | MIF | In vivo: MIF KO →Increased fibrosis in UUO In vivo: MIF inhibition ISO-1 →Increased fibrosis in UUO | In vivo: MIF KO → Increased fibrosis and lower SAM recruitment in CCl4 In vitro: MIF Inhibition ISO-1 → Decreased fibrosis in hepatitis B virus-related liver fibrosis | In vivo: MIF KO → Increased fibrosis in Ang-II | In vivo: Anti-MIF → reduced mortality, no reduction in fibrosis In vivo: ISO-1 and 31 → Decreased fibrosis In vivo: SIX-1 KO → decreased fibrosis | ? |
| D-DT | In vivo: D-DT KO → Increased fibrosis in UUO | ? | Cardiomyocyte- specific D-DT KO: Increased fibrosis | ? | ? | |
| Functional role: Upregulation | MIF | In vivo: Administration recombinant MIF → Decreased fibrosis in UUO | In vivo: Administration recombinant MIF → Decreased fibrosis | In vitro: Recombinant MIF → Decreased fibrosis or increased fibrosis | ? | In vitro: MIF increased the resistance to apoptosis by inhibiting sodium nitroprusside- induced apoptosis in dermal fibroblasts |
| D-DT | In vivo: Administration recombinant D-DT → Decreased fibrosis in UUO | ? | In vitro: Recombinant D-DT→ Decreased fibrosis | ? | ? | |
| Functional role: Mechanism | MIF | Tubulointerstitial fibrosis: MIF abrogates cell cycle arrest of tubular epithelial cells | TAA: Decreasing TGF-α1, PDGF-BB, MMP2, MMP9, TIMP-1 CCl4: CD74/AMPK MIF dependent SAM recruitment through MMP13 In vitro: NASH: CD74/AMPK In vitro: Hepatitis B virus-induced: MIF enhanced the TGF-β/SMAD through CD74 | In vitro: Myofibroblast →Activation CD74, TGF-β2, MMP-2 →MIF/CD74: Antifibrotic →MIF/CXCR4: Profibrotic | In vitro: Lung fibroblasts →MIF stimulate proliferation | In vitro: Dermal fibroblasts decrease the apoptosis through activation of ERK, AKT and Bcl-2 signaling |
| D-DT | Acute kidney injury (I/R): tubular cell regeneration through SLPI- and ATF4-dependent mechanisms | CCl4: Acceleration of the melanin biosynthesis, leading to the protection of the liver from oxidative stress | In vitro: Myofibroblast →inhibiting TGF-β induced SMAD-2 activation CD74/CaMKK2 | ? | ? | |
| Anti-/Profibrotic | MIF | In vivo: Antifibrotic in tubulointerstitial fibrosis | In vivo: Antifibrotic in liver fibrosis; In vitro: Hepatoprotection in NASH | In vivo: Antifibrotic In vitro: Anti-/Profibrotic | In vivo: Antifibrotic | In vitro: Potentially Profibrotic |
| D-DT | In vivo: Antifibrotic in tubulointerstitial fibrosis | In vivo: Potentially antifibrotic | In vivo/In vitro: Antifibrotic | ? | ? | |
2.2.1. Kidney fibrosis
Kidney fibrosis is the terminal pathway of almost all kidney diseases (81). It is the key pathological process behind chronic kidney disease (CKD), which is defined as a chronic dysfunction or structural abnormality of the kidney independent of the cause (82). Worldwide, more than 10% of the population is affected by CKD, which is associated with high morbidity and mortality (83). The three main kidney compartments can all be affected by various diseases and lead to fibrosis (81). Fibrosis in each compartment is termed differently, i.e., tubulointerstitial fibrosis in the tubulointerstitium, glomerulosclerosis in the glomeruli, and arteriosclerosis in the vasculature (27). With disease progression, fibrosis can extend to other compartments; for instance, glomerular diseases can initially lead to glomerulosclerosis and subsequently to tubulointerstitial fibrosis (81). We focus on the role of MIF and D-DT in tubulointerstitial fibrosis and glomerulosclerosis, as there are no studies available evaluating MIF and D-DT in renal arteriosclerosis.
In healthy human and murine kidneys, MIF was mainly expressed in the renal tubular epithelial, endothelial, mesangial cells, and podocytes (84, 85). In human kidneys with tubulointerstitial fibrosis, MIF expression was significantly decreased (84). In contrast, MIF was significantly elevated in the serum of patients with CKD compared with healthy individuals (86). Interestingly, the authors found no correlation between glomerular filtration rate and MIF serum concentration but a correlation with markers of oxidative stress and endothelial activation. In patients with focal glomerular sclerosis, levels of MIF in the urine were significantly higher than those of healthy individuals and correlated with the extent of mesangial matrix increase and that of tubulointerstitial fibrosis. (87) Systemic lupus erythematosus (SLE) is a serious autoimmune disease that can often lead to kidney fibrosis. Increased MIF concentration in the serum, kidneys, and urine have been correlated with active systemic disease as well (88).
Various animal models are used to study kidney diseases, and many develop kidney fibrosis (81). For tubulointerstitial fibrosis, animal models like unilateral ureteral obstruction (UUO) or ischemia-reperfusion (I/R) were used. MIF was less expressed in kidney fibrosis induced by UUO or I/R (84). This reflects the expression in humans, as MIF is less expressed in patients’ samples of kidney fibrosis (84). Nothing is known about the expression of MIF or D-DT in glomerulosclerosis. However, there are findings about MIF and D-DT in the context of glomerular diseases like IgA nephropathy or rapidly progressive glomerulonephritis, which can lead to glomerulosclerosis. Here, a higher MIF expression was found in the glomeruli. A murine model of crescentic glomerulonephritis reflected the situation in humans. (86) MIF was secreted from podocytes, parietal epithelial cells, and mesangial cells in this murine model (85).
The functional role of MIF in tubulointerstitial fibrosis was investigated by UUO and I/R in a systemic MIF knockout (KO). Significantly more fibrosis formation was observed in MIF KO compared to wild-type mice. In an in vivo intervention study, the MIF inhibitor (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) was administered therapeutically three days after fibrosis induction by UUO. The inhibitor significantly increased fibrosis and inflammation. Administration of recombinant MIF resulted in reduced fibrosis and inflammation. In vitro, deletion of MIF in LPS-stimulated tubular epithelial cells leads to increased G2/M cell cycle arrest and increased expression of CDK inhibitor 1B and proinflammatory and profibrotic mediators. Thereby, MIF abrogated cell cycle arrest of tubular epithelial cells, induced apoptosis, and thereby reduced kidney fibrosis. (84)
Autosomal dominant polycystic kidney disease is characterized by renal cyst formation, inflammation, and fibrosis. MIF promotes cystic epithelial cell proliferation by activating ERK, mTOR, and Rb/E2F pathways. MIF also regulated cystic renal epithelial cell apoptosis through p53-dependent signaling. The effect on fibrosis was not directly investigated. (89)
No study investigated the functional role of MIF in glomerulosclerosis. However, one study investigated the connection between MIF and glomerulonephritis, which leads to glomerulosclerosis. Therefore, they used a murine model of crescentic glomerulonephritis, showing that MIF is upregulated and mediates glomerular injury through MIF and its receptor complex CD74/CD44. (85)
Initial data shows that D-DT is mainly located in proximal tubular epithelial cells in healthy human and murine kidneys. In human and murine fibrotic kidneys, D-DT was downregulated. (90) The functional role of D-DT was studied in acute kidney injury. Acute kidney injury can progress to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis (78). D-DT was suggested to mediate proximal tubular cell regeneration via secretory leukocyte protease inhibitor- and activating transcription factor 4-dependent mechanisms in acute kidney injury (22). Initial data indicated increased fibrosis in D-DT KO mice and a potential protective role of D-DT in the UUO model of kidney fibrosis (90).
In the kidney, MIF-family seems to have antifibrotic effects in tubulointerstitial fibrosis. However, many kidney diseases, particularly glomerular diseases, involve complex processes with extensive cellular interactions in different kidney compartments. MIF appears to be disease-promoting in immune and autoimmune-mediated glomerulonephritis and may therefore contribute to secondary glomerulosclerosis, which in turn might drive tertial tubulointerstitial fibrosis. Likely, the role of MIF in kidney fibrosis depends on the specific disease and its stage.
2.2.2. Liver fibrosis
Liver fibrosis is caused by nearly every chronic liver injury or inflammation, e.g., alcohol consumption, non-alcoholic steatohepatitis, chronic viral hepatitis, autoimmune hepatitis, Schistosoma mansoni infection, etc. Mechanistically, activation of hepatic stellate cells, which in turn acquire a myofibroblast-like phenotype, leads to liver fibrosis. In this manner, tissue remodeling in the liver is initiated through the secretion of ECM proteins and MMPs by activated hepatic stellate cells. (91) Compared to other organs, the liver has a high regenerative capacity. With respect to terminology, the most advanced stage of liver fibrosis is termed cirrhosis. Cirrhosis is defined as the histological development of regenerative nodules surrounded by fibrous bands, representing advanced chronic injury and organ failure, associated with high mortality. (92)
MIF was expressed in hepatocytes and hepatic stellate cells (93, 94). Impaired survival was shown in patients with decompensated cirrhosis having high MIF and low sCD74 serum concentrations compared to controls. The increase in MIF serum concentration was independent of liver function but reflected systemic inflammation. It was hypothesized that sCD74 would neutralize MIF in the systemic circulation and specifically antagonize its proinflammatory function. (9) In autoimmune liver diseases such as primary biliary cholangitis and autoimmune hepatitis, serum MIF concentration and hepatic MIF expression were elevated compared to healthy controls (95).
Liver fibrosis can be evaluated using various in vivo models. A distinction was made between chemical, dietary, surgical, transgenic, and immune liver fibrosis models (96). The functional role of MIF in liver fibrosis was investigated in two in vivo chemical, toxin models using carbon tetrachloride (CCl4) and thioacetamide (80, 93, 97). CCl4 led to oxidative damage caused by lipid peroxidation (98). Thioacetamide-induced liver injury is mainly caused by reaction metabolites secreted by thioacetamide, which activate hepatic stellate cells and produce fibrinogen and growth factors (96).
In CCl4-induced liver fibrosis, MIF KO showed increased fibrosis. It was associated with alterations in fibrosis-relevant genes but not by a changed intrahepatic immune cell infiltration. The administration of recombinant MIF displayed decreased fibrosis by CD74/AMPK. (80) Another study focused on the recruitment of scar-associated macrophages by MIF in CCl4-induced liver fibrosis, since scar-associated macrophages regulate hepatic stellate cell activation and ECM degradation. Recruitment of scar-associated macrophages was lower in MIF KO mice compared with wild-type mice. MIF-dependent recruitment of scar-associated macrophages facilitates ECM degradation through MMP13, emphasizing the crucial role of proper macrophage recruitment and phenotypic characteristics in fibrosis resolution. (93) In a thioacetamide-induced liver fibrosis model, MIF showed an antifibrotic activity by decreasing TGF-β1, platelet-derived growth factor-BB, MMP2, MMP9, and tissue inhibitors of MMP1 (97).
In an in vitro nonalcoholic fatty liver disease model, hepatocytes were stimulated with oleic acid. MIF showed a protective effect in hepatocytes by reducing the triglyceride content through CD74/AMPK signaling. Blockade of CD74 but not CXCR2 or CXCR4 fully reverted the protective effect of MIF, comparable to AMPK inhibition. (94) Since non-alcoholic steatohepatitis is one important cause of liver fibrosis, an antifibrotic and protective role of MIF in this disease could be assumed in hepatocytes.
Chronic hepatitis B and C virus infection is a major risk factor for liver fibrosis, cirrhosis, and hepatocellular carcinoma. In a hepatitis B virus-induced liver fibrosis in vitro model, hepatic stellate cells were stimulated with a hepatitis B virus-containing cell culture supernatant and the hepatitis B virus surface antigen. Hepatitis B virus-induced upregulation of MIF and CD74 in hepatic stellate cells. MIF expression correlated with the activation of fibrosis markers, and MIF inhibition with ISO-1, attenuated fibrosis markers. The authors identified MIF to enhance the TGF-β/SMAD signaling through its receptor CD74, promoting hepatic stellate cell activation. Besides, MIF participates in a positive feedback loop, where TGF-β1 further upregulates MIF expression. (48) In hepatitis C virus-induced liver fibrosis, MIF polymorphisms are associated with disease severity and complications in a stage- and context-dependent manner (99).
Schistosoma mansoni infection invariably results in liver fibrosis (100). In patients with a low burden of these parasites, a positive association was found between plasma concentrations of MIF and markers of liver fibrosis (101).
In summary, this could indicate that MIF has an antifibrotic effect in toxin-induced liver fibrosis, but a profibrotic effect in hepatitis B-induced fibrosis.
In an in vivo CCl4 rodent model, an increased production of D-DT was detected. D-DT increase led to an acceleration of melanin biosynthesis, leading to the protection of the liver from oxidative stress. They introduced D-DT as a candidate key protein protecting the rodent liver from CCl4 damage. (102) In a follow-up study, an N-terminal proline glutathionyl carbonylate modification of D-DT was detected, which was formed by condensation of phosgene and reduced glutathione in damaged CCl4 livers (103). Albeit fibrosis was not analyzed, it could be expected that the improved liver damage will be accompanied by reduced liver fibrosis.
Taken together, the role of MIF can differ in liver fibrosis based on the disease leading to fibrosis, with its signaling pathways showing contrasting pro- or antifibrotic effects (48). Currently, there is no study evaluating the direct effect of D-DT in liver fibrosis. In a liver injury model involving liver fibrosis, a protective mechanism induced by D-DT could be identified.
2.2.3. Heart fibrosis
Cardiovascular diseases are a leading cause of mortality worldwide. Heart fibrosis is an essential component and consequence of cardiovascular diseases. It can be divided into two major forms: replacement and reactive (interstitial) fibrosis. Replacement fibrosis occurs focally after necrosis of cardiomyocytes due to ischemia. (104) Heart fibrosis is a pathological process, but it is important to prevent rupture of the heart after myocardial infarction (105). Interstitial fibrosis occurs diffusely in the myocardium as a result of various diseases, e.g., hypertension or diabetes (104). Both forms of heart fibrosis lead to systolic and diastolic dysfunction as well as arrhythmias (106).
In healthy rats, MIF was constitutively expressed at low levels by cardiomyocytes. MIF expression was rapidly upregulated in acute myocardial infarction by surviving cardiomyocytes in the infarcted area. (107) MIF expression was also increased in cardiac fibroblasts after ischemia or pressure overload due to transverse aortic constriction (108, 109). MIF is expressed by inflammatory cells and cardiomyocytes in pathological foci of experimental autoimmune myocarditis (110).
The functional role of MIF in heart fibrosis was investigated in an angiotensin-II infusion-induced hypertrophy model, which mimics hypertension-related interstitial heart fibrosis. MIF KO aggravated heart fibrosis and caused decreased systolic function and increased heart weight (111). These in vivo findings align with the in vitro findings, where overexpression of MIF inhibits the expression of fibrosis markers in mouse cardiac fibroblasts by activating CD74 and targeting the profibrotic genes of transforming growth factor beta-2 and MMP-2 (111). In contrast, another study claimed that cardiac fibroblasts in vitro treated with recombinant MIF promoted proliferation and upregulation of fibrosis markers (96). However, the specific activated receptor was not examined, leaving the involvement of CD74 unresolved. The receptors activated by MIF could explain the contradictory pro- and antifibrotic functions. CD74 was described as mediating antifibrotic properties, while CXCR4 promotes profibrotic effects in cardiac myofibroblasts (112). Furthermore, as autoimmune myocarditis can lead to heart fibrosis, the role of MIF in this condition will be discussed (113). Anti-MIF-antibody treatment in both the early and late phases of experimental autoimmune myocarditis significantly inhibited the disease in a vascular cell adhesion protein-1-dependent manner, by suppressing the expression of TNF-α and IL-1β in the heart (110).
MIF was investigated in the context of aging-induced cardiac anomalies, examining the cardiac geometry, contractile and intracellular Ca2+ properties in young (3-4 months) or old (24 months) wild-type and MIF KO mice. The hearts of aged MIF KO mice had higher heart mass, heart-to-body weight ratio, cardiomyocyte size, and interstitial fibrosis compared to those of aged wild-type mice. (114)
Explanted human hearts had significantly reduced D-DT compared to normal or hypertrophic hearts. No difference was evident in D-DT expression between normal and hypertrophic hearts. In mice, cardiac D-DT was increased due to pressure overload seven weeks after transverse aortic constriction, when left ventricular function was still normal. (109)
The functional role of intrinsic cardiomyocyte-derived D-DT was investigated in modulating the cardiac response to left ventricular pressure overload by transverse aortic constriction. Increased interstitial fibrosis was evident in cardiomyocyte-specific D-DT KO hearts one week after transverse aortic constriction. Seven weeks after transverse aortic constriction, cardiomyocyte-specific D-DT KO mice continued to exhibit a significantly higher degree of fibrosis. Recombinant D-DT in vitro exhibited an antifibrotic effect in cardiac fibroblasts by inhibiting TGF-β-induced SMAD-2 activation. Cardiomyocyte-specific MIF KO demonstrated little susceptibility to heart failure, even when extending the observation period to 10 weeks after transverse aortic constriction. Myocardial D-DT expression was mildly increased in cardiomyocyte-specific MIF KO and might have had a compensatory effect in preventing a phenotype. In contrast, cardiomyocyte-specific D-DT KO mice showed no compensatory increase in MIF after transverse aortic constriction. (109) In a previous study, a protective effect of D-DT required activation of the metabolic stress enzyme AMPK, which was mediated by a CD74/CaMKK2-dependent mechanism after ischemic injury (115).
In summary, an antifibrotic effect of MIF triggered by CD74 was found in heart fibrosis. It is noteworthy that MIF appears crucial for maintaining physiological homeostasis in the heart, as aged MIF KO mice showed heart fibrosis. One study showed a potential profibrotic effect of MIF that could be triggered by CXCR4. D-DT has an antifibrotic and compensatory effect in heart fibrosis, probably mediated by a CD74/CaMKK2-dependent mechanism. All functional studies focused on interstitial heart fibrosis, not replacement fibrosis.
2.2.4. Pulmonary fibrosis
Pulmonary fibrosis is the predominant component of all interstitial lung diseases. Many patients with pulmonary fibrosis can be diagnosed with an autoimmune disorder or hypersensitivity pneumonitis. In patients where no identifiable cause can be determined, the diagnosis of idiopathic pulmonary fibrosis (IPF) is proposed. IPF is a life-threatening disease leading to progressive chronic hypoxemic respiratory failure, and only very limited treatment options are available. It is suggested that a chronic alveolar epithelium injury leads to the activation of fibroblasts, followed by an accumulation of ECM. The injured alveolar epithelium is characterized by cellular senescence and secretion of profibrotic mediators. Immune cells, the microbiome, and alterations in mucin expression are also supposed to be involved in the pathogenesis of IPF. (116) Fibrotic processes are involved in other lung diseases such as cystic fibrosis (CF). CF is an autosomal-recessive, monogenetic disorder caused by mutations in the CF transmembrane conductance regulator gene (117). Because of the reduced mucociliary clearance, patients with CF suffer from recurrent pulmonary bacterial infections (e.g., caused by Pseudomonas aeruginosa or Haemophilus influenzae), causing airway remodeling and finally irreversible loss of lung function. (118)
In the lung, MIF was expressed in the bronchiolar and alveolar epithelium (119). In IPF, MIF was additionally expressed in fibrotic areas, especially in their peripheral regions (119, 120). In bronchoalveolar lavage of patients diagnosed with IPF, a higher MIF concentration was detected compared to healthy individuals (120). In serum, MIF concentration was associated with a higher 3-month mortality in patients with acute exacerbation of IPF (121). CF patients carrying at least one 5-CATT repeat allele exhibit low MIF expression and experience a favorable lung function outcome compared to those without the 5-CATT repeat allele, who are high MIF expressers (122).
Immune cells might be involved in the pathogenesis of IPF. Peripheral lymphocytes from patients with IPF produced more MIF when stimulated with collagen I compared to lymphocytes from healthy individuals (19, 123). Single-cell transcriptomics of lung tissue from patients with IPF revealed that MIF was also involved in the interaction between IPF-associated macrophages and other macrophage subpopulations (124), albeit the functional relevance of MIF in this interaction remains unclear.
A model of pulmonary fibrosis induced by the intratracheal application of bleomycin is widely used (46, 125, 126). As is often the case, this animal model only partially replicates IPF, e.g., it is not inherently idiopathic (116). In this model in mice revealed an increased MIF concentration in the bronchoalveolar lavage, and increased MIF expression in fibrotic areas (46, 125, 126). This is consistent with the findings in patients with IPF as described above.
The functional role of MIF in this animal model has been investigated in several studies. Using a neutralizing anti-MIF antibody, mice showed reduced mortality and infiltration with immune cells but no reduction in fibrosis (125). Using the mouse model and two specific MIF inhibitors ISO-1 and N-(3-hydroxy-4-fluorobenzyl)-5 trifluoromethylbenzoxazol-2-thione 31, another study was able to demonstrate an antifibrotic effect of MIF inhibition (126). The anti-inflammatory effect and a reduction in pulmonary hypertension were also shown (126). A third study using this model in mice confirmed the antifibrotic role of MIF inhibition. They knocked down a MIF transcription factor sine oculis homeobox homolog 1, which is upregulated in alveolar type II cells of lungs with IPF and in the mouse model and drives MIF expression in these cells. The authors further proved in vitro that MIF could stimulate the proliferation of primary human lung fibroblasts. (127) In rats with the bleomycin model, MIF inhibition via a lentivirus reduced lung injury, inflammation, and fibrosis (46, 126). The authors suggested this is driven by the TGF-β1/SMADs signaling pathway (46). An essential process in pulmonary fibrosis is vascular remodeling. MIF inhibition might protect against this process via thrombospondin 2 and serpin family B member 5 signaling (46).
In summary, there is growing evidence suggesting a protective effect of MIF inhibition in pulmonary fibrosis and IPF. So far, nothing is known about the role of D-DT in pulmonary fibrosis.
2.2.5. Systemic sclerosis (Scleroderma)
Systemic sclerosis is a rare autoimmune connective-tissue disease divided into subtypes, such as limited or diffuse cutaneous systemic sclerosis (128, 129). Systemic sclerosis has the highest mortality among all rheumatic diseases (128). It is characterized by progressive fibrosis of the skin and internal organs (129). The most affected organs are the lungs, pericardium, kidneys, skeletal muscle, and gastrointestinal tract (128). The pathogenesis of systemic sclerosis is not completely understood. Several processes are proposed, including microvascular injury, inflammatory cell infiltration, excessive proliferation of fibroblasts, and resistance of fibroblasts to apoptosis. (130) Over time, excessive collagen deposition thickens the skin and restricts joint movement, leading to contractures and disability. As several organs are affected and various complications occur, the treatment is complex, and the progression of the disease can only be slowed down, but not completely stopped. (128) We will focus on the role of MIF in the skin in systemic sclerosis.
Serum levels of MIF were significantly higher in patients with systemic sclerosis than in healthy controls (130–132). However, in another larger study, MIF serum levels showed no difference between systemic sclerosis patients and healthy controls. MIF mRNA expression was significantly lower in peripheral blood mononuclear cells in systemic sclerosis patients compared to controls, while CD74 showed no difference. Immunohistochemistry staining of healthy skin revealed a strong MIF expression in all layers of the epidermis, in endothelium, and in fibroblasts of the papillary dermis. (133) In skin biopsies of systemic sclerosis patients, MIF was enhanced in keratinocytes, dermal fibroblasts, and decreased in endothelial cells (130, 132, 133). MIF expression in diffuse systemic sclerosis patients’ skin was detected in suprabasal keratinocytes, perivascular clusters of infiltrating mononuclear cells, and fibroblast-like cells (132).
To date, the functional role of MIF in systemic sclerosis has not been investigated in vivo. However, in vitro, the functional role of MIF was investigated in systemic sclerosis-derived dermal fibroblasts, which showed a lower degree of apoptosis compared to normal fibroblasts (130, 132). Even when using the apoptosis inducer sodium nitroprusside, systemic sclerosis-derived dermal fibroblasts showed a lower degree of apoptosis than normal dermal fibroblasts. MIF further increased the resistance to apoptosis by inhibiting sodium nitroprusside-induced apoptosis in dermal fibroblasts in a dose-dependent manner. Both ERK inhibition and AKT inhibition almost completely blocked the inhibitory effect of MIF on apoptosis in dermal fibroblasts. (130) These in vitro data indicated that MIF might enhance fibrosis in systemic sclerosis by increasing the resistance to apoptosis of dermal fibroblasts.
The serum concentration of D-DT showed no significant differences between systemic sclerosis patients and control patients. However, this study showed significantly higher D-DT serum concentration in systemic sclerosis patients with a low forced vital capacity of the lungs compared to those without. (131) No further data on D-DT in systemic sclerosis are available.
In summary, several studies described and correlated MIF in serum in systemic sclerosis. However, little functional data exist, overall suggesting disease aggravation by MIF.
2.3. Interventional clinical trials
Several interventional clinical trials using MIF or CD74 inhibition have been conducted, largely in the cancer field. Albeit none of them were performed in fibrotic diseases per se, we provide a brief overview of these trials showing the various available drugs for potential use in patients (based on a search on ClinicalTrials.gov). To date, there was no trial with drugs targeting D-DT or D-DTL.
Imalumab, also known as BAX69, is a recombinant, human monoclonal antibody that specifically binds to a β-sheet structure within oxMIF, including a highly conserved catalytic motif (57Cys-Ala-Leu-Cys60) (134). So far, Imalumab has only been tested in malignant tumors. One study investigated an unspecified anti-MIF antibody in lupus nephritis, but this study was terminated, and no results were published (NCT01541670) (Table 2).
Table 2. Overview of the clinical studies in which MIF, oxMIF, or CD74 was used as a target.
| ID | Phase | Target | Drug | Condition | Status |
|---|---|---|---|---|---|
| NCT01765790 | 1 | oxMIF | Imalumab (BAX69) | Metastatic adenocarcinoma of the colon or rectum, malignant solid tumors | Completed |
| NCT02448810 | 2 | oxMIF | Imalumab (BAX69) | Metastatic colorectal cancer | Terminated |
| NCT02540356 | 1,2 | oxMIF | Imalumab (BAX69) | Refractory ovarian cancer with recurrent symptomatic malignant ascite | Terminated |
| NCT06212076 | 1,2 | MIF | IPG1094 | Solid tumor | Not yet recruiting |
| NCT05112159 | 1 | MIF | IPG1094 | Safety issues | Unknown status |
| NCT01541670 | 1 | MIF | unknown | Systemic lupus erythematosus | Terminated |
| NCT03782415 | 1,2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Glioblastoma, recurrent glioblastoma | Active, not recruiting |
| NCT03489850 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Alcohol use disorder | Completed |
| NCT02025998 | 1 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Alcohol use disorder | Completed |
| NCT02714036 | 1,2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Amyotrophic lateral sclerosis | Completed |
| NCT01317992 | 1,2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Medication overuse headache | Unknown status |
| NCT01860807 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Methamphetamine dependence | Completed |
| NCT01389193 | 1 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Migraine headache | Completed |
| NCT04429555 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Pneumonia, viral (covid-19) | Completed |
| NCT03489850 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Alcohol use disorder | Completed |
| NCT03533387 | 1 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Healthy volunteers | Completed |
| NCT04054206 | 1 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Healthy volunteers | Completed |
| NCT01217970 | 1 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Methamphetamine-dependence, substance abuse, | Completed |
| NCT04057898 | 2, 3 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Amyotrophic lateral sclerosis | Completed |
| NCT02238626 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Amyotrophic lateral sclerosis | Completed |
| NCT05513560 | 2, 3 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Long COVID | Active, not recruiting |
| NCT01982942 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Progressive multiple sclerosis | Completed |
| NCT03594435 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Alcohol use disorder | Completed |
| NCT05414240 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Alcohol use disorder | Recruiting |
| NCT03341078 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Methamphetamine dependence | Recruiting |
| NCT01740414 | 2 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Opioid abuse | Completed |
| NCT04631471 | 3 | MIF, PDE3, 4, 10 | Ibudilast (MN-166) | Myelopathy, spinal cord diseases | Recruiting |
| NCT01663766 | 1 | CD74 | Milatuzumab (hLL1, IMMU-115) | GVHD (acute or chronic) | Terminated |
| NCT00421525 | 1, 2 | CD74 | Milatuzumab (hLL1, IMMU-115) | Multiple myeloma | Completed |
| NCT00504972 | 1 | CD74 | Milatuzumab (hLL1, IMMU-115) | Non-hodgkin’s lymphoma, chronic lymphocytic leukemia | Completed |
| NCT00868478 | 1, 2 | CD74 | Milatuzumab (hLL1, IMMU-115) | Chronic lymphocytic leukemia | Unknown status |
| NCT00989586 | 1, 2 | CD74 | Milatuzumab (hLL1, IMMU-115) | Relapsed and refractory B-cell NHL | Completed |
| NCT01845740 | 1 | CD74 | Milatuzumab (hLL1, IMMU-115) | Systemic lupus erythematosus | Completed |
| NCT00603668 | 1, 2 | CD74 | Milatuzumab (hLL1, IMMU-115) | Chronic lymphocytic lymphoma | Completed |
| NCT01101594 | 1, 2 | ADC (CD74) (Conjugat with Doxorubicin) | hLL1-DOX | Multiple myeloma | Terminated |
| NCT01585688 | 1, 2 | ADC (CD74) (Conjugat with Doxorubicin) | hLL1-DOX | Non-hodgkin’s lymphoma, chronic lymphocytic leukemia | Terminated |
| NCT05611853 | 1,2 | ADC (CD74) | BN301 | Advanced B-cell malignancies | Terminated |
| NCT06563804 | 1, 2 | ADC (CD74) | S227928 | Relapsed/refractory AML, MDS/AML, CMML | Not recruiting |
| NCT03424603 | 1 | ADC (CD74) | STRO-001 | Advanced B-cell malignancies | Completed |
Ibudilast is a small-molecule inhibitor of MIF. It was first discovered as an inhibitor of phosphodiesterase 3, 4, and 10, and secondarily, an MIF-inhibiting function was described (135). Ibudilast binds as a noncompetitive inhibitor to the enzymatically active site of MIF (135). This active site is closely related to the binding site of MIF to CD74 (136). This prevents MIF from binding to CD74 (136). Ibudilast has so far been studied in tumor diseases and neuronal and psychiatric diseases, but not yet in fibrotic diseases (Table 2). IPG1094 is another small-molecule inhibitor of MIF, which is under investigation in two trials in healthy individuals and patients with advanced solid tumors (Table 2).
CD74 has been targeted by the monoclonal antibody Milatuzumab, also known as hLL1 or IMMU-115. Six trials investigated Milatuzumab in hematological neoplasms. Milatuzumab was investigated in patients with systemic lupus erythematosus (SLE), showing no safety concerns and reduced disease activity (NCT01845740) (137). Five trials investigate anti-CD74-antibody-drug-conjugates (ADC) in hematological malignancies (Table 2).
3. Conclusion and future prospects
The MIF-family has emerged as a crucial regulator of tissue remodeling in organ fibrosis. In in vivo fibrosis models, MIF showed predominantly antifibrotic effects, except for lung fibrosis. This is interesting since inflammation is mostly considered to drive fibrosis and MIF is considered pro-inflammatory. D-DT showed antifibrotic effects in fibrosis, albeit much less data is available compared to MIF. The role of D-DTL remains unexplored. Another open question is whether and how the MIF-family members might functionally interact in fibrosis.
MIF and D-DT lead to proliferation through signaling pathways such as ERK signaling. In the kidney, the proliferation of tubular cells exerts regenerative effects and is proposed as the main mechanism of anti-fibrotic effects in the kidneys. In lung fibrosis, the profibrotic effects were related to the proliferation of fibroblasts.
MIF has already been tested in clinical studies for various diseases, with no clear positive effects reported so far (Table 2). Trials in fibrotic diseases are missing. Given the potentially opposing effects of MIF in various diseases and organs, a highly selective patient population might be required for such trials. Also, given the different effects on various cells, future research should focus on specific targeting of such treatments. This is particularly true for fibrosis, which would normally require prolonged therapeutic interventions, thereby elevating the risk of adverse effects. The potential of D-DT as a therapeutic target is currently unclear. Its slightly different receptor-binding profile with potentially lower proinflammatory actions could have beneficial effects on some diseases, but dedicated studies are needed.
While many open questions remain, the MIF-family remains an interesting molecular system involved in the pathophysiology of organ fibrosis. Ultimately, a comprehensive understanding of the MIF-family has the potential to improve the way we diagnose, monitor, and treat fibrotic diseases.
Acknowledgments
Graphical abstract created with BioRender. Boor, P. (2025) https://BioRender.com/qvtlt9y
Abbreviations
- ADC
Antibody-drug-conjugates
- AKI
Acute kidney injury
- AKT
Protein kinase B
- AMPK
AMP-activated protein kinase
- bp
Base pairs
- CD
Cluster of differentiation
- CKD
Chronic kidney disease
- CF
Cystic fibrosis
- Col
Collagen
- CXCR
CXC motif chemokine receptors
- D-DT
D-dopachrome tautomerase
- D-DTL
D-dopachrome tautomerase like
- ECM
Extracellular matrix
- ERK
Extracellular signal-regulated kinase
- IL
Interleukin
- I/R
Ischemia-reperfusion
- IPF
Idiopathic pulmonary fibrosis
- kDa
Kilodalton
- KO
Knockout
- LPS
Lipopolysaccharide
- MIF
Macrophage-migration-inhibitory-factor
- MMP
Matrix metalloproteinase
- NF-κB
Nuclear factor-κB
- OxMIF
Oxidized MIF
- PDGF
Platelet-derived growth factor
- RedMIF
Reduced MIF
- sCD74
soluble CD74
- TGF-β
Transforming growth factor-β
- TNF
Tumor necrosis factor
- UUO
Unilateral ureteral obstruction
Footnotes
Disclosures
The authors report no conflicts of interest.
Grants
PB is supported by the German Research Foundation (DFG, Project IDs 322900939 & 445703531), European Research Council (ERC Consolidator Grant No 101001791), and the Federal Ministry of Education and Research (BMBF, STOP-FSGS-01GM2202C). PD is supported by the START-Program of the Faculty of Medicine RWTH Aachen University (grant number 102/24).
References
- 1.Zhang JM, An J. Cytokines, Inflammation and Pain. Int Anesthesiol Clin. 2007;45:27. doi: 10.1097/AIA.0b013e318034194e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2014;1843:2563–2582. doi: 10.1016/j.bbamcr.2014.05.014. [DOI] [PubMed] [Google Scholar]
- 3.Bloom BR, Bennett B. Mechanism of a reaction in vitro associated with delayed-type hypersensitivity. Science. 1966;153:80–82. doi: 10.1126/science.153.3731.80. [DOI] [PubMed] [Google Scholar]
- 4.David JR. Delayed hypersensitivity in vitro: its mediation by cell-free substances formed by lymphoid cell-antigen interaction. Proc Natl Acad Sci U S A. 1966;56:72–77. doi: 10.1073/pnas.56.1.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Odh G, Hindemith A, Rosengren AM, Rosengren E, Rorsman H. Isolation of a new tautomerase monitored by the conversion of D-dopachrome to 5,6-dihydroxyindole. Biochem Biophys Res Commun. 1993;197:619–624. doi: 10.1006/bbrc.1993.2524. [DOI] [PubMed] [Google Scholar]
- 6.Sugimoto H, Taniguchi M, Nakagawa A, Tanaka I, Suzuki M, Nishihira J. Crystal structure of human D-dopachrome tautomerase, a homologue of macrophage migration inhibitory factor, at 1.54 A resolution. Biochemistry. 1999;38:3268–3279. doi: 10.1021/bi982184o. [DOI] [PubMed] [Google Scholar]
- 7.Leng L, Metz CN, Fang Y, Xu J, Donnelly S, Baugh J, Delohery T, Chen Y, Mitchell RA, Bucala R. MIF Signal Transduction Initiated by Binding to CD74. J Exp Med. 2003;197:1467. doi: 10.1084/jem.20030286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ralchev Ralchev N, Lyubenova Bradyanova S, Valerieva Doneva Y, Mihaylova N, Vikentieva Elefterova-Florova E, Ivanov Tchorbanov A, Munoz-Valle JF, Petralia MC, Checconi P, Nicoletti F, Fagone P. Exploring the Role of CD74 and D-Dopachrome Tautomerase in COVID-19: Insights from Transcriptomic and Serum Analyses. J Clin Med. 2023;12:5037. doi: 10.3390/jcm12155037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wirtz TH, Reuken PA, Jansen C, Fischer P, Bergmann I, Backhaus C, Emontzpohl C, Reißing J, Brandt EF, Koenen MT, Schneider KM, et al. Balance between macrophage migration inhibitory factor and sCD74 predicts outcome in patients with acute decompensation of cirrhosis. JHEP Reports. 2021;3:100221. doi: 10.1016/j.jhepr.2020.100221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Tanese K, Ogata D. The role of macrophage migration inhibitory factor family and CD74 in the pathogenesis of melanoma. Exp Dermatol. 2024;33:e15122. doi: 10.1111/exd.15122. [DOI] [PubMed] [Google Scholar]
- 11.Westmeier J, Brochtrup A, Paniskaki K, Karakoese Z, Werner T, Sutter K, Dolff S, Limmer A, Mittermüller D, Liu J, Zheng X, et al. Macrophage migration inhibitory factor receptor CD74 expression is associated with expansion and differentiation of effector T cells in COVID-19 patients. Front Immunol. 2023;14:1236374. doi: 10.3389/fimmu.2023.1236374. /FULL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Weber C, Kraemer S, Drechsler M, Lue H, Koenen RR, Kapurniotu A, Zernecke A, Bernhagen J. Structural determinants of MIF functions in CXCR2-mediated inflammatory and atherogenic leukocyte recruitment. Proc Natl Acad Sci U S A. 2008;105:16278–16283. doi: 10.1073/pnas.0804017105. /SUPPL_FILE/0804017105SI.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Alampour-Rajabi S, El Bounkari O, Rot A, Müller-Newen G, Bachelerie F, Gawaz M, Weber C, Schober A, Bernhagen J. MIF interacts with CXCR7 to promote receptor internalization, ERK1/2 and ZAP-70 signaling, and lymphocyte chemotaxis. FASEB Journal. 2015;29:4497–4511. doi: 10.1096/fj.15-273904. [DOI] [PubMed] [Google Scholar]
- 14.Lourenco S, Teixeira VH, Kalber T, Jose RJ, Floto RA, Janes SM. Macrophage Migration Inhibitory Factor - CXCR4 is the dominant chemotactic axis in human mesenchymal stem cell recruitment to tumors. J Immunol. 2015;194:3463. doi: 10.4049/jimmunol.1402097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mangano K, Diamantopoulos A, Vallianou NG, Stratigou T, Panagopoulos F, Kounatidis D, Dalamaga M, Fagone P, Nicoletti F. Serum and urinary levels of MIF, CD74, DDT and CXCR4 among patients with type 1 diabetes mellitus, type 2 diabetes and healthy individuals: Implications for further research. Metabol Open. 2024;24:100320. doi: 10.1016/j.metop.2024.100320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Song S, Liu B, Habibie H, van den Bor J, Smit MJ, Gosens R, Wu X, Brandsma CA, Cool RH, Haisma HJ, Poelarends GJ, et al. D-dopachrome tautomerase contributes to lung epithelial repair via atypical chemokine receptor 3-dependent Akt signaling. EBioMedicine. 2021;68 doi: 10.1016/j.ebiom.2021.103412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang S, Gao S, Li Y, Qian X, Luan J, Lv X. Emerging Importance of Chemokine Receptor CXCR4 and Its Ligand in Liver Disease. Front Cell Dev Biol. 2021;9:716842. doi: 10.3389/fcell.2021.716842. /BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zan C, Yang B, Brandhofer M, El Bounkari O, Bernhagen J. D-dopachrome tautomerase in cardiovascular and inflammatory diseases—A new kid on the block or just another MIF? The FASEB Journal. 2022;36:e22601. doi: 10.1096/fj.202201213R. [DOI] [PubMed] [Google Scholar]
- 19.Florez-Sampedro L, Soto-Gamez A, Poelarends GJ, Melgert BN. The role of MIF in chronic lung diseases: Looking beyond inflammation. Am J Physiol Lung Cell Mol Physiol. 2020;318:L1183–L1197. doi: 10.1152/ajplung.00521.2019. /ASSET/IMAGES/LARGE/ZH50062078290007.JPEG. [DOI] [PubMed] [Google Scholar]
- 20.Benedek G, Meza-Romero R, Jordan K, Zhang Y, Nguyen H, Kent G, Li J, Siu E, Frazer J, Piecychna M, Sreih A, et al. MIF and D-DT are potential disease severity modifiers in male MS subjects. Proc Natl Acad Sci U S A. 2017;114:E8421–E8429. doi: 10.1073/pnas.1712288114. /-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Rajasekaran D, Zierow S, Syed M, Bucala R, Bhandari V, Lolis EJ. Targeting distinct tautomerase sites of D-DT and MIF with a single molecule for inhibition of neutrophil lung recruitment. The FASEB Journal. 2014;28:4961. doi: 10.1096/fj.14-256636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ochi A, Chen D, Schulte W, Leng L, Moeckel N, Piecychna M, Averdunk L, Stoppe C, Bucala R, Moeckel G. MIF-2/D-DT enhances proximal tubular cell regeneration through SLPI- and ATF4-dependent mechanisms. Am J Physiol Renal Physiol. 2017;313:F767. doi: 10.1152/ajprenal.00683.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bounkari E, El Bounkari O, Zan C, Wagner J, Bugar E, Bourilhon P, Kontos C, Zarwel M, Sinitski D, Milic J, Jansen Y, et al. MIF-2/D-DT is an atypical atherogenic chemokine that promotes advanced atherosclerosis and hepatic lipogenesis. doi: 10.1038/s41467-025-57540-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mutsaers HAM, Merrild C, Nørregaard R, Plana-Ripoll O. The impact of fibrotic diseases on global mortality from 1990 to 2019. J Transl Med. 2023;21:1–3. doi: 10.1186/s12967-023-04690-7. /FIGURES/1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Abbafati C, Abbas KM, Abbasi M, Abbasifard M, Abbasi-Kangevari M, Abbastabar H, Abd-Allah F, Abdelalim A, Abdollahi M, Abdollahpour I, Abedi A, et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. The Lancet. 2020;396:1204–1222. doi: 10.1016/S0140-6736(20)30925-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Djudjaj S, Boor P. Cellular and molecular mechanisms of kidney fibrosis. Mol Aspects Med. 2019;65:16–36. doi: 10.1016/j.mam.2018.06.002. [DOI] [PubMed] [Google Scholar]
- 28.Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nature Reviews Molecular Cell Biology. 2024;25(8):617–638. doi: 10.1038/s41580-024-00716-0. [DOI] [PubMed] [Google Scholar]
- 29.Boor P, Ostendorf T, Floege J. Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nature Reviews Nephrology. 2010;6(11):643–656. doi: 10.1038/nrneph.2010.120. [DOI] [PubMed] [Google Scholar]
- 30.Merk M, Mitchell RA, Endres S, Bucala R. D-dopachrome tautomerase 1 (D-DT or MIF-2): Doubling the MIF cytokine family. Cytokine. 2012;59:10. doi: 10.1016/j.cyto.2012.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Esumi N, Budarf M, Ciccarelli L, Sellinger B, Kozak CA, Wistow G. Conserved gene structure and genomic linkage for D-dopachrome tautomerase (DDT) and MIF. Mamm Genome. 1998;9:753–757. doi: 10.1007/s003359900858. [DOI] [PubMed] [Google Scholar]
- 32.Song S, Xiao Z, Dekker FJ, Poelarends GJ, Melgert BN. Macrophage migration inhibitory factor family proteins are multitasking cytokines in tissue injury. Cell Mol Life Sci. 2022;79:105. doi: 10.1007/s00018-021-04038-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rosengren E, Åman P, Thelin S, Hansson C, Ahlfors S, Björk P, Jacobsson L, Rorsman H. The macrophage migration inhibitory factor MIF is a phenylpyruvate tautomerase. FEBS Lett. 1997;417:85–88. doi: 10.1016/s0014-5793(97)01261-1. [DOI] [PubMed] [Google Scholar]
- 34.Senter PD, Al-Abed Y, Metz CN, Benigni F, Mitchell RA, Chesney J, Han J, Gartner CG, Nelson SD, Todaro GJ, Bucala R. Inhibition of macrophage migration inhibitory factor (MIF) tautomerase and biological activities by acetaminophen metabolites. Proc Natl Acad Sci U S A. 2002;99:144–149. doi: 10.1073/pnas.011569399. /ASSET/01ED3147-3826-4DCA-98CA-97B7138D0C38/ASSETS/GRAPHIC/PQ2615693004.JPEG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fingerle-Rowson G, Kaleswarapu DR, Schlander C, Kabgani N, Brocks T, Reinart N, Busch R, Schütz A, Lue H, Liu A, Xiong H, et al. A tautomerase-null macrophage migration-inhibitory factor (MIF) gene knock-in mouse model reveals that protein interactions and not enzymatic activity mediate MIF-dependent growth regulation. Mol Cell Biol. 2009;29:1922–1932. doi: 10.1128/MCB.01907-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tilstam PV, Pantouris G, Corman M, Andreoli M, Mahboubi K, Davis G, Leng L, Lolis E, Bucala R. A selective small-molecule inhibitor of macrophage migration inhibitory factor-2 (MIF-2), a MIF cytokine superfamily member, inhibits MIF-2 biological activity. J Biol Chem. 2019;294:18522. doi: 10.1074/jbc.RA119.009860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Grieb G, Merk M, Bernhagen J, Bucala R. Macrophage migration inhibitory factor (MIF): a promising biomarker. Drug News Perspect. 2010;23:257. doi: 10.1358/dnp.2010.23.4.1453629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schober A, Bernhagen J, Weber C. Chemokine-like functions of MIF in atherosclerosis. J Mol Med (Berl) 2008;86:761–770. doi: 10.1007/s00109-008-0334-2. [DOI] [PubMed] [Google Scholar]
- 39.Merk M, Zierow S, Leng L, Das R, Schulte W, Fan J, Lue H, Chen Y, Xiong H, Chagnon F, Bernhagen J, et al. The D-dopachrome tautomerase (DDT) gene product is a cytokine and functional homolog of macrophage migration inhibitory factor (MIF) Proc Natl Acad Sci U S A. 2011;108:E577–E585. doi: 10.1073/pnas.1102941108. /ASSET/909FBD7D-2E62-4334-BC2F-9091CBEA8D7D/ASSETS/GRAPHIC/PNAS.1102941108FIG06.JPEG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Calandra T, Bernhagen J, Mitchell RA, Bucala R. The macrophage is an important and previously unrecognized source of macrophage migration inhibitory factor. J Exp Med. 1994;179:1895–1902. doi: 10.1084/jem.179.6.1895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mitchell RA, Liao H, Chesney J, Fingerle-Rowsor G, Baugh J, David J, Bucala R. Macrophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: regulatory role in the innate immune response. Proc Natl Acad Sci U S A. 2002;99:345–350. doi: 10.1073/pnas.012511599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rajasekaran D, Gröning S, Schmitz C, Zierow S, Drucker N, Bakou M, Kohl K, Mertens A, Lue H, Weber C, Xiao A, et al. Macrophage Migration Inhibitory Factor-CXCR4 Receptor Interactions: EVIDENCE FOR PARTIAL ALLOSTERIC AGONISM IN COMPARISON WITH CXCL12 CHEMOKINE. J Biol Chem. 2016;291:15881–15895. doi: 10.1074/jbc.M116.717751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Meza-Romero R, Benedek G, Jordan K, Leng L, Pantouris G, Lolis E, Bucala R, Vandenbark AA. Modeling of both shared and distinct interactions between MIF and its homologue D-DT with their common receptor CD74. Cytokine. 2016;88:62–70. doi: 10.1016/j.cyto.2016.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, Kooistra T, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007;13:587–596. doi: 10.1038/nm1567. [DOI] [PubMed] [Google Scholar]
- 45.Calandra T, Roger T. Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol. 2003;3:791. doi: 10.1038/nri1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Luo Y, Yi H, Huang X, Lin G, Kuang Y, Guo Y, Xie C. Inhibition of macrophage migration inhibitory factor (MIF) as a therapeutic target in bleomycin-induced pulmonary fibrosis rats. Am J Physiol Lung Cell Mol Physiol. 2021;321:L6–L16. doi: 10.1152/ajplung.00288.2020. /ASSET/IMAGES/LARGE/AJPLUNG.00288.2020_F0 06.JPEG. [DOI] [PubMed] [Google Scholar]
- 47.Yu X, Lin SG, Huang XR, Bacher M, Leng L, Bucala R, Lan HY. Macrophage Migration Inhibitory Factor Induces MMP-9 Expression in Macrophages via The MEK-ERK MAP Kinase Pathway. 2007;27:103–109. doi: 10.1089/jir.2006.0054. https://home.liebertpub.com/jir . [DOI] [PubMed] [Google Scholar]
- 48.Cheng JY, Shan GY, Wan H, Zhang YX, Gao ZC, Shi YP, Liu F, Yan W qun, Li HJ. MIF/CD74 axis in hepatic stellate cells mediates HBV-related liver fibrosis. Int Immunopharmacol. 2025;147:113929. doi: 10.1016/j.intimp.2024.113929. [DOI] [PubMed] [Google Scholar]
- 49.Thiele M, Donnelly SC, Mitchell RA. OxMIF: a druggable isoform of macrophage migration inhibitory factor in cancer and inflammatory diseases. J Immunother Cancer. 2022;10:e005475. doi: 10.1136/jitc-2022-005475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mahalingam D, Patel MR, Sachdev JC, Hart LL, Halama N, Ramanathan RK, Sarantopoulos J, Liu X, Yazji S, Jäger D, Yoon M, et al. Anti-oxidized macrophage migration inhibitory factor (oxMIF) antibody imalumab (BAX69) in advanced solid tumors: Final results of first-in-human phase 1 study. Annals of Oncology. 2016;27:vi116. doi: 10.1093/annonc/mdw368.08. [DOI] [Google Scholar]
- 51.Chen E, Reiss K, Shah D, Manjula R, Allen B, Murphy EL, Murphy JW, Batista VS, Bhandari V, Lolis EJ, Lisi GP. A structurally preserved allosteric site in the MIF superfamily affects enzymatic activity and CD74 activation in D-dopachrome tautomerase. Journal of Biological Chemistry. 2021;297 doi: 10.1016/j.jbc.2021.101061. /ATTACHMENT/80B8E7B9-3DA2-4540-A669-E6603C4162DE/MMC1.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Chen E, Widjaja V, Kyro G, Allen B, Das P, Prahaladan VM, Bhandari V, Lolis EJ, Batista VS, Lisi GP. Mapping N-to C-terminal allosteric coupling through disruption of a putative CD74 activation site in D-dopachrome tautomerase. J Biol Chem. 2023;299 doi: 10.1016/j.jbc.2023.104729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Parkins A, Pilien AVR, Wolff AM, Argueta C, Vargas J, Sadeghi S, Franz AH, Thompson MC, Pantouris G. The C-terminal Region of D-DT Regulates Molecular Recognition for Protein-Ligand Complexes. J Med Chem. 2024;67:7359–7372. doi: 10.1021/acs.jmedchem.4c00177. [DOI] [PubMed] [Google Scholar]
- 54.Illescas O, Pacheco-Fernández T, Laclette JP, Rodriguez T, Rodriguez-Sosa M. Immune modulation by the macrophage migration inhibitory factor (MIF) family: D-dopachrome tautomerase (DDT) is not (always) a backup system. Cytokine. 2020;133:155121. doi: 10.1016/j.cyto.2020.155121. [DOI] [PubMed] [Google Scholar]
- 55.Ssadh H Al, Spencer PS, Alabdulmenaim W, Alghamdi R, Madar IH, Miranda-Sayago JM, Fernández N, Al Ssadh H, Spencer PS, Alabdulmenaim W, Alghamdi R, et al. Measurements of heterotypic associations between cluster of differentiation CD74 and CD44 in human breast cancer-derived cells. Oncotarget. 2017;8:92143–92156. doi: 10.18632/oncotarget.20922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Farr L, Ghosh S, Moonah S. Role of MIF Cytokine/CD74 Receptor Pathway in Protecting Against Injury and Promoting Repair. Front Immunol. 2020;11:543730. doi: 10.3389/fimmu.2020.01273. /BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Shi X, Leng L, Wang T, Wang W, Li J, McDonald C, Chen Z, Murphy JW, Lolis E, Noble P, Knudson W, et al. CD44 Is the Signaling Component of the Macrophage Migration Inhibitory Factor-CD74 Receptor Complex. Immunity. 2006;25:595. doi: 10.1016/j.immuni.2006.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Leng L, Metz CN, Fang Y, Xu J, Donnelly S, Baugh J, Delohery T, Chen Y, Mitchell RA, Bucala R. MIF Signal Transduction Initiated by Binding to CD74. J Exp Med. 2003;197:1467. doi: 10.1084/JEM.20030286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Huntington JT, Shields JM, Wyatt CA, Benbow U, Slingluff CL, Brinckerhoff CE. Overexpression of collagenase 1 (MMP-1) is mediated by the ERK pathway in invasive melanoma cells. Role of BRAF mutation and fibroblast growth factor signaling. Journal of Biological Chemistry. 2004;279:33168–33176. doi: 10.1074/jbc.M405102200. /ASSET/8F92AE5A-9836-4F79-B228-0050DEE9F129/MAIN.ASSETS/GR7.JPG. [DOI] [PubMed] [Google Scholar]
- 60.Becker-Herman S, Arie G, Medvedovsky H, Kerem A, Shachar I. CD74 Is a Member of the Regulated Intramembrane Proteolysis-processed Protein Family. Mol Biol Cell. 2005;16:5061. doi: 10.1091/mbc.E05-04-0327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Parkins A, Chen E, Rangel VM, Singh M, Xue L, Lisi GP, Pantouris G. Ligand-induced conformational changes enable intersubunit communications in D-dopachrome tautomerase. Biophys J. 2023;122:1268. doi: 10.1016/j.bpj.2023.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hoffmann A, Bernhagen J. Revisiting the secretion mechanism(s) of macrophage migration inhibitory factor—welcome to the “UPS club”. Immunol Cell Biol. 2020;98:704–708. doi: 10.1111/imcb.12388. [DOI] [PubMed] [Google Scholar]
- 63.Merk M, Baugh J, Zierow S, Leng L, Pal U, Lee SJ, Ebert AD, Mizue Y, Trent JO, Mitchell R, Nickel W, et al. The Golgi-Associated Protein p115 Mediates the Secretion of Macrophage Migration Inhibitory Factor. The Journal of Immunology. 2009;182:6896–6906. doi: 10.4049/jimmunol.0803710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Brocks T, Fedorchenko O, Schliermann N, Stein A, Moll UM, Seegobin S, Dewor M, Hallek M, Marquardt Y, Fietkau K, Heise R, et al. Macrophage migration inhibitory factor protects from nonmelanoma epidermal tumors by regulating the number of antigen-presenting cells in skin. The FASEB Journal. 2016;31:526. doi: 10.1096/fj.201600860R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Abe R, Shimizu T, Ohkawara A, Nishihira J. Enhancement of macrophage migration inhibitory factor (MIF) expression in injured epidermis and cultured fibroblasts. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2000;1500:1–9. doi: 10.1016/s0925-4439(99)00080-0. [DOI] [PubMed] [Google Scholar]
- 66.Cheng Q, McKeown SJ, Santos L, Santiago FS, Khachigian LM, Morand EF, Hickey MJ. Macrophage Migration Inhibitory Factor Increases Leukocyte–Endothelial Interactions in Human Endothelial Cells via Promotion of Expression of Adhesion Molecules. The Journal of Immunology. 2010;185:1238–1247. doi: 10.4049/jimmunol.0904104. [DOI] [PubMed] [Google Scholar]
- 67.Fingerle-Rowson GR, Bucala R. Neuroendocrine properties of macrophage migration inhibitory factor (MIF) Immunol Cell Biol. 2001;79:368–375. doi: 10.1046/j.1440-1711.2001.01024.x. [DOI] [PubMed] [Google Scholar]
- 68.Zhang Y, Zhu W, He H, Fan B, Deng R, Hong Y, Liang X, Zhao H, Li X, Zhang F. Macrophage migration inhibitory factor rejuvenates aged human mesenchymal stem cells and improves myocardial repair. Aging. 2019;11:12641–12660. doi: 10.18632/aging.102592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.He XX, Yang J, Ding YW, Liu W, Shen QY, Xia HHX. Increased epithelial and serum expression of macrophage migration inhibitory factor (MIF) in gastric cancer: potential role of MIF in gastric carcinogenesis. Gut. 2006;55:797–802. doi: 10.1136/gut.2005.078113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kim DHJ, Iosif AM, Ramirez-Celis A, Ashwood P, Ames JL, Lyall K, Berger K, Croen LA, Van de Water J. Neonatal immune signatures differ by sex regardless of neurodevelopmental disorder status: macrophage migration inhibitory factor (MIF) alone reveals a sex by diagnosis interaction effect. Brain Behav Immun. 2023;111:328. doi: 10.1016/j.bbi.2023.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Aloisi AM, Pari G, Ceccarelli I, Vecchi I, Ietta F, Lodi L, Paulesu L. Gender-related effects of chronic non-malignant pain and opioid therapy on plasma levels of macrophage migration inhibitory factor (MIF) Pain. 2005;115:142–151. doi: 10.1016/j.pain.2005.02.019. [DOI] [PubMed] [Google Scholar]
- 72.The Human Protein Atlas. 2025. Mar 1, [Online]. [date unknown] https://www.proteinatlas.org/
- 73.Schröder B. The multifaceted roles of the invariant chain CD74 — More than just a chaperone. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2016;1863:1269–1281. doi: 10.1016/j.bbamcr.2016.03.026. [DOI] [PubMed] [Google Scholar]
- 74.Noer JB, Talman MLM, Moreira JMA. HLA class II histocompatibility antigen γ chain (CD74) expression is associated with immune cell infiltration and favorable outcome in breast cancer. Cancers (Basel) 2021;13:6179. doi: 10.3390/cancers13246179. /S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gao Y, Li J, Cheng W, Diao T, Liu H, Bo Y, Liu C, Zhou W, Chen M, Zhang Y, Liu Z, et al. Cross-tissue human fibroblast atlas reveals myofibroblast subtypes with distinct roles in immune modulation. Cancer Cell. 2024;42:1764–1783.:e10. doi: 10.1016/j.ccell.2024.08.020. [DOI] [PubMed] [Google Scholar]
- 76.Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R, Murimwa G, Wright S, Gu X, Maddipati R, Müller S, et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell. 2022;40:656. doi: 10.1016/j.ccell.2022.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang L, Woltering I, Holzner M, Brandhofer M, Schaefer CC, Bushati G, Ebert S, Yang B, Muenchhoff M, Hellmuth JC, Scherer C, et al. CD74 is a functional MIF receptor on activated CD4+ T cells. Cellular and Molecular Life Sciences. 2024;81:1–24. doi: 10.1007/s00018-024-05338-5. /METRICS. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol. 2024;327:F788–F805. doi: 10.1152/ajprenal.00184.2024. /ASSET/IMAGES/LARGE/AJPRENAL.00184.2024_F001.JPEG. [DOI] [PubMed] [Google Scholar]
- 79.Zhang Y, Lu S, Fan S, Xu L, Jiang X, Wang K, Cai B. Macrophage migration inhibitory factor activates the inflammatory response in joint capsule fibroblasts following post-traumatic joint contracture. Aging (Albany NY) 2021;13:5804. doi: 10.18632/aging.202505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Heinrichs D, Knauel M, Offermanns C, Berres ML, Nellen A, Leng L, Schmitz P, Bucala R, Trautwein C, Weber C, Bernhagen J, et al. Macrophage migration inhibitory factor (MIF) exerts antifibrotic effects in experimental liver fibrosis via CD74. Proc Natl Acad Sci U S A. 2011;108:17444–17449. doi: 10.1073/pnas.1107023108. /SUPPL_FILE/PNAS.201107023SI.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol. 2024;327:F788–F805. doi: 10.1152/AJPRENAL.00184.2024. /ASSET/IMAGES/LARGE/AJPRENAL.00184.2024_F001.JPEG. [DOI] [PubMed] [Google Scholar]
- 82.Stevens PE, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancıoğlu R, Lamb E, et al. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int. 2024;105:S117–S314. doi: 10.1016/j.kint.2023.10.018. [DOI] [PubMed] [Google Scholar]
- 83.Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl (2011) 2022;12:7. doi: 10.1016/j.kisu.2021.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Djudjaj S, Martin IV, Buhl EM, Nothofer NJ, Leng L, Piecychna M, Floege J, Bernhagen J, Bucala R, Boor P. Macrophage migration inhibitory factor limits renal inflammation and fibrosis by counteracting tubular cell cycle arrest. Journal of the American Society of Nephrology. 2017;28:3590–3604. doi: 10.1681/ASN.2017020190. /-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Djudjaj S, Lue H, Rong S, Papasotiriou M, Klinkhammer BM, Zok S, Klaener O, Braun GS, Lindenmeyer MT, Cohen CD, Bucala R, et al. Macrophage migration inhibitory factor mediates proliferative GN via CD74. Journal of the American Society of Nephrology. 2016;27:1650–1664. doi: 10.1681/ASN.2015020149. /-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bruchfeld A, Carrero JJ, Qureshi AR, Lindholm B, Barany P, Heimburger O, Hu M, Lin X, Stenvinkel P, Miller EJ. Elevated Serum Macrophage Migration Inhibitory Factor (MIF) Concentrations in Chronic Kidney Disease (CKD) Are Associated with Markers of Oxidative Stress and Endothelial Activation. Molecular Medicine. 2008;15:70. doi: 10.2119/molmed.2008.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Matsumoto K, Maruyama N, Maruyama T, Ohnishi Y, Nonaka S, Inoshita A, Ito K, Kitajima S, Abe M, Satomura A, Fujita T. Elevated macrophage migration inhibitory factor (MIF) levels in the urine of patients with focal glomerular sclerosis. Clin Exp Immunol. 2005;139:338. doi: 10.1111/j.1365-2249.2004.02670.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bilsborrow JB, Doherty E, Tilstam PV, Bucala R. Macrophage migration inhibitory factor (MIF) as a therapeutic target for rheumatoid arthritis and systemic lupus erythematosus. Expert Opin Ther Targets. 2019;23:733. doi: 10.1080/14728222.2019.1656718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen L, Zhou X, Fan LX, Yao Y, Swenson-Fields KI, Gadjeva M, Wallace DP, Peters DJM, Yu A, Grantham JJ, Li X. Macrophage migration inhibitory factor promotes cyst growth in polycystic kidney disease. J Clin Invest. 2015;125:2399. doi: 10.1172/JCI80467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Herkens L, Droste P, Karyniotakis K, Boor P, Djudjaj S. The Role of D-dopachrome Tautomerase (Macrophage Migration Inhibitory Factor 2) in Kidney Fibrosis. Journal of the American Society of Nephrology. 2024;35 doi: 10.1681/ASN.2024PSTH7B5P. [DOI] [Google Scholar]
- 91.Aydin MM, Akcali KC. Liver fibrosis. The Turkish Journal of Gastroenterology. 2018;29:14. doi: 10.5152/tjg.2018.17330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Schuppan D, Afdhal NH. Liver Cirrhosis. Lancet. 2008;371:838. doi: 10.1016/S0140-6736(08)60383-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Barnes MA, McMullen MR, Roychowdhury S, Madhun NZ, Niese K, Olman MA, Stavitsky AB, Bucala R, Nagy LE. Macrophage migration inhibitory factor is required for recruitment of scar-associated macrophages during liver fibrosis. J Leukoc Biol. 2014;97:161. doi: 10.1189/jlb.3A0614-280R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Heinrichs D, Berres ML, Coeuru M, Knauel M, Nellen A, Fischer P, Philippeit C, Bucala R, Trautwein C, Wasmuth HE, Bernhagen J. Protective role of macrophage migration inhibitory factor in nonalcoholic steatohepatitis. The FASEB Journal. 2014;28:5136–5147. doi: 10.1096/fj.14-256776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Assis DN, Leng L, Zhang CK, Grieb G, Merk M, Garcia AB, Mccrann C, Chapiro J, Meinhardt A, Mizue Y, Nikolic-Paterson DJ, et al. The role of macrophage migration inhibitory factor in autoimmune liver disease. Hepatology. 2014;59:580–591. doi: 10.1002/hep.26664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wu ST, Wang XX, Xing WB, Li FY, Liang M, Li KS, He Y, Wang JM. An update on animal models of liver fibrosis. Front Med (Lausanne) 2023;10:1160053. doi: 10.3389/fmed.2023.1160053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Radosavljevic T, Vukicevic D, Djuretic J, Gopcevic K, Labudovic Borovic M, Stankovic S, Samardzic J, Radosavljevic M, Vucevic D, Jakovljevic V. The Role of Macrophage Inhibitory Factor in TAA-Induced Liver Fibrosis in Mice: Modulatory Effects of Betaine. Biomedicines. 2024;12:1337. doi: 10.3390/biomedicines12061337. /S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bao YL, Wang L, Pan HT, Zhang TR, Chen YH, Xu SJ, Mao XL, Li SW. Animal and Organoid Models of Liver Fibrosis. Front Physiol. 2021;12:666138. doi: 10.3389/fphys.2021.666138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wirtz TH, Fischer P, Backhaus C, Bergmann I, Brandt EF, Heinrichs D, Koenen MT, Schneider KM, Eggermann T, Kurth I, Stoppe C, et al. Genetic Variants in the Promoter Region of the Macrophage Migration Inhibitory Factor are Associated with the Severity of Hepatitis C Virus-Induced Liver Fibrosis. Int J Mol Sci. 2019;20:3753. doi: 10.3390/ijms20153753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Andrade ZA. Schistosomiasis and liver fibrosis. Parasite Immunol. 2009;31:656–663. doi: 10.1111/j.1365-3024.2009.01157.x. [DOI] [PubMed] [Google Scholar]
- 101.Rodrigues Oliveira JL, Teixeira MM, Lambertucci JR, Antunes CMF, Carneiro M, Negrão-Corrêa D. Plasma levels of innate immune mediators are associated with liver fibrosis in low parasite burden Schistosoma mansoni-infected individuals. Scand J Immunol. 2018;87:e12642. doi: 10.1111/sji.12642. [DOI] [PubMed] [Google Scholar]
- 102.Hiyoshi M, Konishi H, Uemura H, Matsuzaki H, Tsukamoto H, Sugimoto R, Takeda H, Dakeshita S, Kitayama A, Takami H, Sawachika F, et al. D-Dopachrome tautomerase is a candidate for key proteins to protect the rat liver damaged by carbon tetrachloride. Toxicology. 2009;255:6–14. doi: 10.1016/j.tox.2008.09.016. [DOI] [PubMed] [Google Scholar]
- 103.Hiyoshi M, Osago H, Kobayashi-Miura M, Hara N. Detection of a glutathionyl-carbonylated group (GS–CO–) on D-dopachrome tautomerase with preferential binding of GS–CO– to MIF proteins in rat livers damaged by carbon tetrachloride. Chem Biol Interact. 2023;374:110406. doi: 10.1016/j.cbi.2023.110406. [DOI] [PubMed] [Google Scholar]
- 104.Hinderer S, Schenke-Layland K. Cardiac fibrosis – A short review of causes and therapeutic strategies. Adv Drug Deliv Rev. 2019;146:77–82. doi: 10.1016/j.addr.2019.05.011. [DOI] [PubMed] [Google Scholar]
- 105.Jiang W, Xiong Y, Li X, Yang Y. Cardiac Fibrosis: Cellular Effectors, Molecular Pathways, and Exosomal Roles. Front Cardiovasc Med. 2021;8 doi: 10.3389/fcvm.2021.715258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Frangogiannis NG. Cardiac fibrosis. Cardiovasc Res. 2021;117:1450–1488. doi: 10.1093/cvr/cvaa324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yu CM, Lai KWH, Chen YX, Huang XR, Lan HY. Expression of macrophage migration inhibitory factor in acute ischemic myocardial injury. Journal of Histochemistry and Cytochemistry. 2003;51:625–631. doi: 10.1177/002215540305100508. /ASSET/IMAGES/LARGE/10.1177_002215540305100508-FIG4.JPEG. [DOI] [PubMed] [Google Scholar]
- 108.Voss S, Krüger S, Scherschel K, Warnke S, Schwarzl M, Schrage B, Girdauskas E, Meyer C, Blankenberg S, Westermann D, Lindner D. Macrophage Migration Inhibitory Factor (MIF) Expression Increases during Myocardial Infarction and Supports Pro-Inflammatory Signaling in Cardiac Fibroblasts. Biomolecules. 2019;9:38. doi: 10.3390/biom9020038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ma Y, Su KN, Pfau D, Rao VS, Wu X, Hu X, Leng L, Piecychna M, Bedi K, Campbell SG, Eichmann A, et al. Cardiomyocyte d-dopachrome tautomerase protects against heart failure. JCI Insight. 2019;4 doi: 10.1172/jci.insight.128900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Matsui Y, Okamoto H, Jia N, Akino M, Uede T, Kitabatake A, Nishihira J. Blockade of macrophage migration inhibitory factor ameliorates experimental autoimmune myocarditis. J Mol Cell Cardiol. 2004;37:557–566. doi: 10.1016/j.yjmcc.2004.05.016. [DOI] [PubMed] [Google Scholar]
- 111.Liang J nan, Zou X, Fang X hong, Xu J dong, Xiao Z, Zhu J ning, Li H, Yang J, Zeng N, Yuan S jing, Pan R, et al. The Smad3-miR-29b/miR-29c axis mediates the protective effect of macrophage migration inhibitory factor against cardiac fibrosis. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2019;1865:2441–2450. doi: 10.1016/j.bbadis.2019.06.004. [DOI] [PubMed] [Google Scholar]
- 112.Soppert J, Kraemer S, Beckers C, Averdunk L, Möllmann J, Denecke B, Goetzenich A, Marx G, Bernhagen J, Stoppe C. Soluble CD74 reroutes MIF/CXCR4/AKT-mediated survival of cardiac myofibroblasts to necroptosis. J Am Heart Assoc. 2018;7 doi: 10.1161/JAHA.118.009384. /SUPPL_FILE/JAH33461-SUP-0001-SUPINFO.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu Y, Zhu H, Su Z, Sun C, Yin J, Yuan H, Sandoghchian S, Jiao Z, Wang S, Xu H. IL-17 contributes to cardiac fibrosis following experimental autoimmune myocarditis by a PKCβ/Erk1/2/NF-κB-dependent signaling pathway. Int Immunol. 2012;24:605–612. doi: 10.1093/intimm/dxs056. [DOI] [PubMed] [Google Scholar]
- 114.Xu X, Pang J, Chen Y, Bucala R, Zhang Y, Ren J. Macrophage Migration Inhibitory Factor (MIF) Deficiency Exacerbates Aging-Induced Cardiac Remodeling and Dysfunction Despite Improved Inflammation: Role of Autophagy Regulation. Sci Rep. 2016;6:22488. doi: 10.1038/srep22488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Qi D, Atsina K, Qu L, Hu X, Wu X, Xu B, Piecychna M, Leng L, Fingerle-Rowson G, Zhang J, Bucala R, et al. The vestigial enzyme D-dopachrome tautomerase protects the heart against ischemic injury. J Clin Invest. 2014;124:3540. doi: 10.1172/JCI73061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lederer DJ, Martinez FJ. Idiopathic Pulmonary Fibrosis. N Engl J Med. 2018;378:1811–1823. doi: 10.1056/NEJMra1705751. [DOI] [PubMed] [Google Scholar]
- 117.Ratjen F, Bell SC, Rowe SM, Goss CH, Quittner AL, Bush A. Cystic fibrosis. Nat Rev Dis Primers. 2015;1:15010. doi: 10.1038/nrdp.2015.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rogers GB, Hart CA, Mason JR, Hughes M, Walshaw MJ, Bruce KD. Bacterial Diversity in Cases of Lung Infection in Cystic Fibrosis Patients: 16S Ribosomal DNA (rDNA) Length Heterogeneity PCR and 16S rDNA Terminal Restriction Fragment Length Polymorphism Profiling. J Clin Microbiol. 2003;41:3548. doi: 10.1128/JCM.41.8.3548-3558.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Olivieri C, Bargagli E, Inghilleri S, Campo I, Cintorino M, Rottoli P. Macrophage migration inhibitory factor in lung tissue of idiopathic pulmonary fibrosis patients. Exp Lung Res. 2016;42:263–266. doi: 10.1080/01902148.2016.1199744. [DOI] [PubMed] [Google Scholar]
- 120.Bargagli E, Olivieri C, Nikiforakis N, Cintorino M, Magi B, Perari MG, Vagaggini C, Spina D, Prasse A, Rottoli P. Analysis of macrophage migration inhibitory factor (MIF) in patients with idiopathic pulmonary fibrosis. Respir Physiol Neurobiol. 2009;167:261–267. doi: 10.1016/j.resp.2009.05.004. [DOI] [PubMed] [Google Scholar]
- 121.Li X, Zhou Y, Zou R, Chen H, Liu X, Qiu X, Xiao Y, Cai H, Dai J. Associations of Serological Biomarkers of sICAM-1, IL-1 β, MIF, and su-PAR with 3-Month Mortality in Acute Exacerbation of Idiopathic Pulmonary Fibrosis. Mediators Inflamm. 2020;2020 doi: 10.1155/2020/4534272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Adamali H, Armstrong ME, McLaughlin AM, Cooke G, McKone E, Costello CM, Gallagher CG, Leng L, Baugh JA, Fingerle-Rowson G, Bucala RJ, et al. Macrophage Migration Inhibitory Factor Enzymatic Activity, Lung Inflammation, and Cystic Fibrosis. Am J Respir Crit Care Med. 2012;186:162. doi: 10.1164/rccm.201110-1864OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kravis TC, Ahmed A, Brown TE, Fulmer JD, Crystal RG. Pathogenic mechanisms in pulmonary fibrosis: collagen-induced migration inhibition factor production and cytotoxicity mediated by lymphocytes. Journal of Clinical Investigation. 1976;58:1223. doi: 10.1172/JCI108576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhang H, Yang Y, Cao Y, Guan J. IPF-related new macrophage subpopulations and diagnostic biomarker identification - combine machine learning with single-cell analysis. Respir Res. 2024;25:1–17. doi: 10.1186/s12931-024-02845-8. /FIGURES/8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Tanino Y, Makita H, Miyamoto K, Betsuyaku T, Ohtsuka Y, Nishihira J, Nishimura M. Role of macrophage migration inhibitory factor in bleomycin-induced lung injury and fibrosis in mice. Am J Physiol Lung Cell Mol Physiol. 2002;283 doi: 10.1152/ajplung.00155.2001. [DOI] [PubMed] [Google Scholar]
- 126.Günther S, Bordenave J, Hua-Huy T, Nicco C, Cumont A, Thuillet R, Tu L, Quatremarre T, Guilbert T, Jalce G, Batteux F, et al. Macrophage Migration Inhibitory Factor (MIF) Inhibition in a Murine Model of Bleomycin-Induced Pulmonary Fibrosis. Int J Mol Sci. 2018;19:4105. doi: 10.3390/ijms19124105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wilson C, Mertens TCJ, Shivshankar P, Bi W, Collum SD, Wareing N, Ko J, Weng T, Naikawadi RP, Wolters PJ, Maire P, et al. Sine oculis homeobox homolog 1 plays a critical role in pulmonary fibrosis. JCI Insight. 2022;7 doi: 10.1172/jci.insight.142984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Volkmann ER, Andréasson K, Smith V. Systemic sclerosis. Lancet. 2022;401:304. doi: 10.1016/S0140-6736(22)01692-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Romanowska-Próchnicka K, Dziewit M, Lesiak A, Reich A, Olesińska M. Scleroderma and scleroderma-like syndromes. Front Immunol. 2024;15:1351675. doi: 10.3389/fimmu.2024.1351675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kim JY, Kwok SK, Hur KH, Kim HJ, Kim NS, Yoo SA, Kim WU, Cho CS. Up-regulated macrophage migration inhibitory factor protects apoptosis of dermal fibroblasts in patients with systemic sclerosis. Clin Exp Immunol. 2008;152:328. doi: 10.1111/j.1365-2249.2008.03637.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Vincent FB, Lin E, Sahhar J, Ngian GS, Kandane-Rathnayake R, Mende R, Hoi AY, Morand EF, Lang T, Harris J. Analysis of serum macrophage migration inhibitory factor and D-dopachrome tautomerase in systemic sclerosis. Clin Transl Immunology. 2018;7:e1042. doi: 10.1002/cti2.1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Selvi E, Tripodi SA, Catenaccio M, Lorenzini S, Chindamo D, Manganelli S, Romagnoli R, Ietta F, Paulesu L, Miracco C, Cintorino M, et al. Expression of macrophage migration inhibitory factor in diffuse systemic sclerosis. Ann Rheum Dis. 2003;62:460. doi: 10.1136/ard.62.5.460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Baños-Hernández CJ, Bucala R, Hernández-Bello J, Navarro-Zarza JE, Villanueva-Pérez MA, Godínez-Rubí M, Parra-Rojas I, Vázquez-Villamar M, Pereira-Suárez AL, Valle JFM. Expression of macrophage migration inhibitory factor and its receptor CD74 in systemic sclerosis. Cent Eur J Immunol. 2021;46:375. doi: 10.5114/ceji.2021.109756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Mahalingam D, Patel MR, Sachdev JC, Hart LL, Halama N, Ramanathan RK, Sarantopoulos J, Völkel D, Youssef A, de Jong FA, Tsimberidou AM. Phase I study of imalumab (BAX69), a fully human recombinant antioxidized macrophage migration inhibitory factor antibody in advanced solid tumours. Br J Clin Pharmacol. 2020;86:1836. doi: 10.1111/bcp.14289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Cho Y, Crichlow GV, Vermeire JJ, Leng L, Du X, Hodsdon ME, Bucala R, Cappello M, Gross M, Gaeta F, Johnson K, Lolis EJ. Allosteric inhibition of macrophage migration inhibitory factor revealed by ibudilast. Proc Natl Acad Sci U S A. 2010;107:11313–11318. doi: 10.1073/pnas.1002716107. /-/DCSUPPLEMENTAL. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Skeens E, Pantouris G, Shah D, Manjula R, Ombrello MJ, Maluf NK, Bhandari V, Lisi GP, Lolis EJ. A Cysteine Variant at an Allosteric Site Alters MIF Dynamics and Biological Function in Homo- and Heterotrimeric Assemblies. Front Mol Biosci. 2022;9 doi: 10.3389/fmolb.2022.783669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Wallace DJ, Figueras F, Wegener WA, Goldenberg DM. Experience with milatuzumab, an anti-CD74 antibody against immunomodulatory macrophage migration inhibitory factor (MIF) receptor, for systemic lupus erythematosus (SLE) Ann Rheum Dis. 2021;80:954–955. doi: 10.1136/annrheumdis-2020-219803. [DOI] [PubMed] [Google Scholar]




