Abstract
Macrophage migration inhibitory factor (MIF) is involved in various immune-mediated pathologies and regulates both innate and adaptive immune reactions, thus being related to several acute and chronic inflammatory diseases such as rheumatoid arthritis, septic shock, and atherosclerosis. Its role in acute and chronic brain pathologies, such as stroke and neurodegenerative diseases, has attracted increasing attention in recent years. In response to stimuli like hypoxia, inflammation or infection, different cell types can rapidly release MIF, including immune cells, endothelial cells, and neuron cells. Notably, clinical data from past decades also suggested a possible link between serum MIF levels and the severity of stroke and the evolving of neurodegenerative diseases. In this review, we summarize the major and recent findings focusing on the mechanisms of MIF modulating functions in brain injury and neurodegenerative diseases, which may provide important therapeutic targets meriting further investigation.
Keywords: Macrophage migration inhibitory factor, inflammatory response, immune reaction, stroke, neurodegenerative disease
Introduction
Macrophage migration inhibitory factor (MIF), a 12.5 kD protein that inhibits macrophage migration, has been primarily recognized as a product of T lymphocytes. 1 Later, rapidly growing evidence revealed that MIF can be broadly expressed in various cell types, including macrophages, smooth muscle cells, endothelial cells, and neuron cells.2–4 The physiopathological roles of MIF have come to light subsequently. It’s an inflammatory cytokine with chemokine-like functions that is involved in several immune-mediated pathologies and is responsible for inflammatory reactions throughout the central nervous system.5–9 The role of MIF in pathological conditions appears to be diverse with both protective and detrimental effects. For example, MIF defective animals showed decreased neuronal death and improved recovery from transient middle cerebral artery occlusion (tMCAo) in the brain, 10 suggesting its detrimental role in acute cerebrovascular diseases.11,12 However, both ischemic and hemorrhagic models could benefit from MIF overexpression by attenuating oxidative stress.13,14 Furthermore, evidence from both clinical studies and basic research have suggested that MIF could be the biomarker to predict the outcome of neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). The purpose of this review is to provide an overview of the research on MIF, with a particular focus on the context of brain injury and neurodegenerative diseases.
Regulatory mechanisms of MIF expression under different pathological contexts
The MIF gene is located on human chromosome 22 (22q11.2) and 10, 15 where functional polymorphisms -794 CATT 5-8 microsatellite repeat (rs5844572) and -173 G > C single nucleotide polymorphism (SNP) (rs755622) control MIF promoter activity and MIF expression. 16 It is expressed at a high level in various parts of the rat brain, including the cortex, hypothalamus, and hippocampus. 16 MIF’s expression can be increased in vivo following physiological and pathological stimuli like hormones, glucose, cytokines, and microbial products. 17 For instance, after lipopolysaccharide (LPS) injection, both MIF mRNA and protein levels were increased and quickly released into the cerebrospinal fluid of the rat, where cytokines such as IL-1β and IL-6 could be detected. 16 A similar elevation of gene expression was also found in a LPS treated marine invertebrate model. 18 What’s more, Rice et al. demonstrated that angiotensin II stimulated MIF mRNA and protein production, which induced a rapid release of 50% of MIF protein within 20 minutes in the rat proximal tubular epithelial cells. 19 MIF, as previously reported, can boost transcription factor nuclear factor kappa B (NF-κB) expression, which therefore has a pro-inflammatory effect, and increasing NF-κB levels triggers MIF gene transcription, creating a feedback loop. However, these effects were context dependent, since NF-κB decreased MIF expression under hypoxia conditions, then diminished the neuronal protective effect of MIF in a cerebral ischemia mouse model. 13 Thus, the regulation of MIF expression is dependent on the different cellular background during physiological or pathological processes.
The intracellular downstream signals of MIF
The MIF homologous receptor, a cluster of differentiation 74 (CD74) was discovered in the early twentieth century. 20 It is a type II transmembrane protein required for MIF-induced mitogen-activated protein kinase (MAPK) cascade activation, cyclooxygenase 2 (COX2) and prostaglandin E2 (PGE2) production, as well as cell proliferation. 20 CD44 or C-X-C Motif containing chemokine receptor 2 (CXCR2), CXCR4, and CXCR7 are examples of co-receptors.21,22 MIF can activate the CD74/CD44 complex via tyrosine-protein kinase (SRC) and phosphorylate downstream MAPK family signals such as ERK1/2, PI3K, and protein kinase B (PKB), also known as AKT.20,23–25 Then, NF-κB and activator protein-1 (AP1) will be activated, 26 resulting in the release of pro-inflammatory cytokines like IL-6, IL-8, IL-10, and TNF-α. 27 Furthermore, MIF functions as a link between NF-κB and the tumor suppressor protein p53, as p53-dependent macrophage apoptosis can be inhibited by MIF, allowing the macrophage to retain its proinflammatory properties. 28 In addition, MIF binding to CXCR2 and CXCR4 is linked to the activation of mononuclear cell chemotaxis, whereas CXCR7 is linked to the ERK- and zeta-chain-associated protein kinase (ZAP)-70 signaling pathways, as well as B-lymphocyte migration.21,22 When CXCR7 and CXCR4 combine to form homo- and heterodimers, they are able to bind CXCL12 with a much more affinity than CXCR4 and subsequently mediate G-protein signaling.29,30 The significance of CXCL12/CXCR4/CXCR7 signaling in prostate cancer of obese mice has been clarified. 31 As a result, it can be assumed that high amounts of MIF may interfere with the CXCL12-CXCR4/CXCR7 axis by competing for binding sites especially after the stroke, 32 and it has been discovered that the MIF/CXCR7/AKT pathway promotes the development and metastasis of castration-resistant prostate cancer. 33
MIF upregulated the expression of toll-like receptor 4 (TLR-4) in response to LPS and Gram-negative bacteria, which was necessary for the production of proinflammatory cytokines such as TNF-α, IL-6, and IL-12.34–36 Moreover, MIF is involved in the inflammatory response of NLR family pyrin domain containing 3 (NLRP3) inflammasome. 37 Macrophages and dendritic cells (DCs) produce less IL-1α, IL-1β and IL-18 when NLRP3 inhibits MIF activation. 38 Hypoxia-inducible factor-1 (HIF-1) increases the transcriptional expression of MIF, and the elevated MIF level prevents HIF-1 degradation by interacting with constitutive photomorphogenesis 9 (COP9) signalosome subunit 5 (CSN5), which stabilizes HIF-1 by inhibiting its prolyl-564 hydroxylation.28,39 What’s more, MIF inhibits the anti-inflammatory and immunosuppressive effects of glucocorticoid (GC) via MAPK phosphatase.40,41 Figure 1 concludes the MIF-mediated immune response signaling transduction.
Figure 1.
The overview of MIF-mediated cellular signaling transduction. MIF binds the ligand-binding protein CD74 and activates MAPK cascade, COX2, and PGE2. MIF activates the CD74/CD44 complex via SRC and phosphorylates downstream ERK1/2, PI3K, and protein kinase B. NF-κB and AP1 then be activated to induce the release of IL-6, IL-8, IL-10, and TNF-α, while the tumor suppressor protein p53 is inhibited by NF-κB. HIF-1 increases the expression of MIF, and the elevated MIF stabilizes HIF-1 in turn by preventing its degradation through CSN5. MIF suppresses GC immunosuppressive effects via MAPK phosphatase and upregulates TLR-4 expression in response to LPS stimuli, then induces the production of downstream proinflammatory cytokines such as TNF-α, IL-6, and IL-12. MIF: macrophage migration inhibitory factor; MAPK: mitogen-activated protein kinase; COX2: cyclooxygenases 2; PGE2: prostaglandin E2; SRC: tyrosine-protein kinase; ERK: extracellular regulated protein kinases; PI3K: Phosphoinositide 3-kinase; AP1: activator protein-1; IL: interleukin; TNF: tumor necrosis factor; HIF-1: Hypoxia-inducible factor-1; CSN5: constitutive photomorphogenesis 9 signalosome subunit 5; GC: glucocorticoid; TLR-4: toll-like receptor 4. The figure were created with BioRender.com.
Divergent pathological mechanisms of MIF in different brain resident cells
Double side effects on neuronal cells
In addition to serving as an immune mediator, MIF has a direct influence on neuronal cells. The protective effects of MIF on neuronal cells via blocking microglial activation were discovered at the end of last century. 42 Sumners et al. then conducted a series of studies using MIF to investigate its function on neurons. 43 They found that MIF, via its intrinsic thiol-protein oxidoreductase (TPOR), acted as an intracellular negative regulator of neuronal activity after the stimulation of Angiotensin II (Ang II), the hormone responsible for increased blood pressure.44–46 This negative regulation was insufficient in spontaneously hypertensive rat neurons, because increased Ang II level did not elevate the expression of MIF and led to the chronotropic effect of Ang II. 47
Zis et al. discovered that the MIF content in cerebral blood vessel endothelial cells was significantly increased in ischemic stroke patients, and that the protective property was also observed in cultured cortical neuron model of oxygen-glucose deprivation (OGD) to mimic ischemia, treatment of MIF for 12 hours prevented OGD-induced cell death. 48 MIF depletion via shRNA in vitro or genetically modified animals was used to examine the effect of MIF on neuron cell death. The absence of MIF increased neuronal cell apoptotic caspase 3/7 activity,13,49 and its presence rescued neurons from death by shifting the pro-/anti-apoptosis pathway balance to the survival side. 50 MIF treatment improved the proliferation and survival of neural stem/progenitor cells via many pathways, including PKB and extracellular regulated protein kinases (Erk), thereby extending these benefits to neural stem/progenitor cells.49,51
In contrast to the positive effects, the detrimental aspects of MIF were also significant. Eric et al. found that after a single injection of 200 ng exogenous MIF into the mouse hippocampus, the pharmacological kinetics of induced N-methyl-D-aspartic acid receptor (NMDA) responses in the apical dendrites of hippocampal CA1 pyramidal neurons were altered for more than two weeks, which may be responsible for sequelae after CNS injury. 52 In another rat diffuse axonal injury model, cortex MIF levels were considerably elevated 3 hours after injury and were primarily localized in neurons, intracerebroventricular injection of the MIF antagonist ISO-1 reduced neuronal apoptosis and axonal injury. 53 The similar neuronal cell death promotional effects of MIF were found to be involved in other studies, including the traumatic brain injury (TBI) model, which led to more severe neurodegeneration and neurologic deficits.10,54
MIF accumulates in astrocytes and regulates its activity
The inhibitory effects of MIF on the reactivity of astrocytes were discovered as early as 1996. 55 In addition, MIF was found to be accumulated in astrocytes at the blood brain barrier (BBB) in the Borna disease virus (BDV) infected rat brain, where it modulates inflammation during virus-induced encephalitis. However, during BDV-induced inflammation, MIF in astrocytes was not produced at the gene level since MIF mRNA was absent in the experimental groups. 56 Recently, Okazaki and colleagues discovered that antipsychotics increased MIF expression in astrocytes via increasing the acetylation of H3K27 in the MIF promoter, indicating that MIF may play a role in the pathophysiology of schizophrenia disease and the working mechanisms of antipsychotics. 57
The promoting effects of MIF on astrocyte activity were investigated at the cellular level as well. Svenningsen et al. found that MIF and Serine protease high-temperature requirement A1 (HTRA1), a tumor suppressor implicated in cell growth and cerebral small vessel disease, 58 were both expressed in cultured mouse astrocytes. 59 HTRA1 can reduce astrocyte migration, whereas MIF inhibits this influence, which has potential benefits for CNS development and disease treatment. 59 The enhanced astrocyte proliferation phenomenon was also observed in a TBI mouse model, MIF concentrations were increased after traumatic brain injury and associated with poor outcome, and the pre-administration of MIF antagonist might prevent TBI induced astrocytosis. 60
MIF produced by microglia and involved in its inflammatory response
Previous studies have suggested that spinal microglia, not invading immune cells, create MIF in inflammatory hyperalgesia.61,62 In primary cultured spinal microglia cells, MIF administration boosted COX2 expression and PGE2 production, while CD74 deletion completely reversed these effects. 63
Overall, the interactions of MIF with microglia are detrimental. The inhibition of MIF tautomerase activity downregulated LPS-induced nitro oxide (NO) and TNF-α production in microglial cells, indicating that MIF is involved in microglia-mediated neuron inflammatory pathology. 64 Zheng et al. found that Z-312, a novel MIF inhibitor, reduced proinflammatory factors produced by LPS-stimulated microglial cells in another LPS-induced PD mouse model. 65 Not surprisingly, MIF induced neuroinflammation and Parkinson's disease-like symptoms when microglial autophagy function was inhibited. 66 In addition to PD, the MIF level in glioblastoma patients was significantly elevated, and its expression in malignant gliomas exceeded that of normal brain tissue. The suppression of the MIF/CD74 pathway promoted interferon (IFN)- secretion in microglia, resulting in the death of tumor cells and a change from M2 to M1 in microglia associated with glioma. 67 Infusion of MIF into the animal hippocampus decreased microglia activation and prevented the production of microglia-derived tissue plasminogen activator, thereby protecting against excitotoxic neuronal cell death. 42
MIF involved in cerebrovascular structural cells dysfunction
Endothelial cells can release and respond to MIF when proatherogenic stimuli are present because they play a crucial structural function in blood vessel walls. Consequently, they can contribute to a number of vascular disorders, including atherosclerosis, retinopathy, and pulmonary hypertension.68–70
MIF was expressed in the cerebral micro-vessels in the peri-infarct area of 10 acute stroke patients, in contrast to negative findings in normal brain. 48 The elevated expression of MIF in both the MCAo and OGD tests led to the discovery that MIF may stimulate angiogenesis via the AKT and ERK signaling pathways in rat brain microvascular endothelial cells. 71 MIF levels in the blood of 39 stroke patients were also shown to be elevated; the subsequent investigation indicated that MIF injection disrupted tight junction in rat OGD-treated brain endothelial cells (ARBECs) and compromised BBB integrity in a tMCAo mice model. 72 In experimental cerebral malaria produced by Plasmodium berghei ANKA, brain endothelial cells that displayed CD74 signaling contributed to the cross-presentation of antigens to CD8+ T lymphocytes, and pharmacologic Plasmodium MIF antagonism protected against cerebral malaria. 73
The mural cells of brain microvasculature is composed of pericytes and vascular smooth muscle cells (VSMCs).74,75 Previous studies demonstrated that human placental NG2+ pericytes are the primary source of MIF in response to pathogenic activation, and these pericytes also offer migratory pathways for extravasated leukocytes in rat mesentery tissue and in vitro EC-PC barrier model.76–78 Human VSMCs are likewise the secretory source of MIF under hypoxic conditions via HIF-1 activation, 79 with vascular remodeling properties through VSMC differentiation and proliferation interference in carotid and pulmonary arteries.80,81 Nevertheless, the interaction of MIF with cerebrovascular pericytes and VSMCs remains limited. Greater comprehension of this topic will allow us to prevent BBB damage and hasten the recovery of the neurovascular unit after a stroke. The interaction of MIF with brain resident cells in experimental stroke were concluded in Table 2.
Table 2.
The dualistic effects of MIF on brain resident cells in experimental stroke.
| Brain cells | Study objects | Major findings | Mechanisms | Time point of main treatment | Duration of brain injury after in vivo model onset |
|---|---|---|---|---|---|
| Neuronal cells | Totoxin-induced hippocampus neurodegeneration 35 | Pretreatment of MIF protect neuronal cell from death | MIF inhibited microglial activation and the subsequent release of tissue plasminogen activator | Two days infusion of 500 μM MIF before the injection of kainate | 5 days |
| Rat hypothalamus–brainstem neuronal cell cultures38,40 | Increased levels of intracellular MIF inhibited the stimulatory actions of Ang II on neuronal firing | The inhibitory actions are mediated via a thiol-oxidoreductase activity of the MIF molecule and subsequent scavenging of ROS | Neuronal cultures were pretreated with Ang II for 5 hours | / | |
| Cortical neurons of E18–E19 rat embryos 41 | MIF significantly protected neurons against OGD-induced cell death and lower cell viability | Not deeply investigated | Simultaneously treated with OGD and 50 or 100 ng/mL human MIF | / | |
| Human neuroblastoma cell line; mice tMCAO model 13 | MIF’s protection of neuronal cell occurs relatively early following stroke (within 10 h) and diminished at late phase (24 h) in vivo | Reduced caspase-3 activation | Simultaneously treated with OGD and human MIF at a concentration of 200 ng/ml in vitro; using MIF-knockout mice in vivo study | 4 and 22 hours | |
| Mice neural stem/progenitor cells 42 | MIF contributed to neural stem/progenitor cells proliferation, survival and migration | MIF increased the phosphorylation of AKT, ERK, AMPK and STAT3; MIF knockdown by shRNA increased caspase 3/7 activity | 3 days treatment of MIF (400 ng/ml) | / | |
| Human neuroblastoma cell line 43 | MIF promoted cell survival in OGD treated neuroblastoma cells | Induced the expression of mature BDNF, MAP2 and Bcl2, and decreased the expression of Caspase-3 and Bax | 30 ng/ml MIF after 4 hours of OGD | / | |
| Kunming mice and hippocampuscells 44 | MIF Is Expressed in NeuN-Positive, NeuN-Negative, and Granule cells in Developing Hippocampus | MIF antagonist suppressed cell proliferation in the developing hippocampus in vivo and hinders neurite outgrowth in vitro | Inject ISO-1 into Postnatal day 5 mice for 9 consecutive days; primary neuronal cultures with ISO-1 using 10 or 100 µM for 2 days | / | |
| Adult male C57BL/6 mice 45 | MIF significantly increased the baseline frequency of Ca2+ events in mouse hippocampal CA1 layer pyramidal neurons | Possibly via an interaction with Kv channels, might directly promoted epileptogenic activity in the hippocampus | Injected 200 ng recombinant MIF into the CA1 region of the hippocampus | 14 days | |
| Rat with diffuse axonal injury (DAI) 46 | MIF was increased and peaked at 1 d after DAI; inhibition of MIF exhibited decreased apoptosis, axonal injury, and glial response | Increased levels of IL-6, IL-1β, and TNF-α through TLR-related pathways following DAI | Injected intracerebroventricularly with 10 μL ISO-1 mixture 0.5 h before modeling | 1 day | |
| TBI mouse model 47 | MIF KO protected dendritic spine density and morphological complexity in neurons and reduced neuronal cell death following TBI | Through MIF nuclease activity rather than pro-infammatory role | MIF KO mice and nuclease-defcient E22A MIF mutant | 1 day | |
| Astrocyte | Adult retired female breeders of the CD1 strain 48 | MIF reduced astroglial reactivity | Not deeply investigated | Single application of 500 μM MIF | 4 days |
| Borna Disease Virus infected rat model 49 | MIF protein accumulation in astrocytes and infifiltration of ED1-positive macrophages | MIF from neurons or other extracellular sources into astrocytes at Blood-Brain Barrier | No specific treatment | 22 days | |
| Astrocytes of P1–P5 mice 51 | MIF rescued HTRA1-inhibited astrocyte migration | The binding between MIF and HTRA1 was to inhibit the proteolytic activity of HTRA1 | Simultaneously treatment of HTRA1 and 50 ng/mL MIF | / | |
| Male C57BL/6J mice with Fluid Percussion Injury (FPI) Model of TBI 52 | ISO-1 reduced astrocyte activation but had no signifificant effffect on neurodegeneration | Not deeply investigated | Injected 10 mg/kg ISO1 at 30 min after TBI | 3 days | |
| Microglia | LPS treated murine microglia cell line and mouse neuroblastoma cell line 56 | MIF Inhibitor Z-590 attenuated the microglia-mediated neurotoxicity to HT-22 neuroblastoma cells | Z-590 decreased the production of NO, TNF-α, IL-6, IL-1β, COX-2, iNOS and ROS involved in inhibiting MAKPs signaling pathway in LPS-stimulated microglia cells | 30 min Z-590 (5–20 μM) prior to and during incubation with LPS (200 ng/mL) or vehicle for an additional 16 h | / |
| LPS treated murine microglia cell line and mouse neuroblastoma cell line, and C57BL/6 mice 57 | MIF Inhibitor Z-312 suppressed the neurotoxic effects in vitro, and ameliorated microglial activation and subsequent DA neuron loss | Z-312 significantly decreased the production of NO, IL-1β, TNF-α and IL-6 | 30 min Z-312 before adding 200 ng/mL of LPS in vitro, intraperitoneally administered Z-312 (20 mg/kg/d) for 10 consecutive days before LPS treatment in vivo | 8 days | |
| Murine glioma and microglia cell lines, rat glioma cell line and human glioma cell line; C57BL/6 mice 59 | The inhibition of MIF or its receptor CD74 promoted IFN-γ release and amplified tumor death as well as inducing a M2 to M1 shift in glioma-associated microglia | MIF-CD74 signaling inhibited interferon (IFN)-γ secretion in microglia through phosphorylation of microglial ERK1/2 | MIF-overexpressing or knockdown and CD74 antibody were used | 5 days | |
| Endothelial cells | Rat with tMCAO and rat brain microvascular endothelial cells 67 | The protective effect of miR-493 inhibition in angiogenesis was attenuated by knocking down MIF | MiR-493 directly modulated MIF expression | Knockout MIF before treatment of miR-493 inhibition | 3 days |
| Rats with tMCAO and pMCAO, and rat brain endothelial cells 68 | MIF increased BBB permeability and exacerbates stroke injury | MIF led to the disruption of tight junction | MIF were given after MCAO (20 µg/kg) or OGD treatment | 4h for permeability; 24 h for infarct volume | |
| Mice infected with Plasmodium berghei ANKA 69 | Cd74−/− mice is resistant to experimental cerebral malaria | Cross-presentation of Plasmodium antigen is MIF/CD74 signaling dependent | Pharmacologic Plasmodium MIF antagonism before infection and continued treatment once daily for 2 days | 7 days |
ERK: extracellular regulated protein kinases; MAPK: mitogen-activated protein kinase; STAT: Signal Transducers and Activators of Transcription; DAI: diffuse axonal injury; BDNF: brain derived neurotrophic factor; MAP2: microtubule association protein-2; FPI: fluid percussion injury; HTRA: high-temperature requirement A1; NO: nitro oxide; TNF: tumor necrosis factor; iNOS: inducible nitric oxide synthase; ROS: reactive oxygen species; BBB: blood brain barrier; LPS: lipopolysaccharide.
The function of MIF on brain injury
Dualistic roles of MIF on ischemic stroke
Although MIF depletion had no effect on cytokine levels in the brain or serum of mice one week after tMCAo, 82 the detrimental effect of MIF on stroke recovery was found elsewhere. 10 All the underlying mechanisms, including pro-inflammation, enhance immune reaction and accelerate neuron cell death, inhibit the recovery of post-stroke physiological function.10,83–85 MIF levels were found to be considerably higher in acute ischemic stroke patients and in the experimental tMCAo model. The serum MIF levels of patients were positively correlated with infarct volume and long-term outcomes, while treatment of MIF increased BBB permeability in animal models following tMCAo.72,86 Acute ischemic stroke patients with higher serum MIF concentrations at admission were more likely to have poor clinical outcome, post-stroke depression (PSD) and recurrent stroke in the long term,87–89 and its major receptor CD74 was also found to have a similar predictive role in ischemic stroke patients. 90 In a different mouse focal ischemia model, knocking down MIF or inhibiting its nuclease activity decreased infarct volume and improved behavioral performance. 91 Based on the nuclease property, the most recent work utilizing a photothrombotic ischemia model demonstrated that MIF acetylation on the K78 residue decreased the translocation and nuclear intensity of MIF, therefore providing a therapeutic target for protecting neurons in ischemic stroke. 92
MIF, on the other hand, has a neuroprotective effect in ischemic stroke by inhibiting neuronal cell death and inducing brain-derived neurotrophic factor (BDNF) expression.12,50,93 It was found that 60 ng/mL MIF administration was most effective for BDNF expression and inhibiting neuronal apoptosis. 94 MIF promoted angiogenesis after ischemic stroke, this process could be inhibited by miR-493, which was previously thought to be a tumor angiogenesis suppressor. 71 Furthermore, the deletion of MIF gene accelerated neuronal loss after stroke because MIF shielded neurons from oxidative stress and ischemia-reperfusion (I/R) induced apoptosis by reducing caspase-3 activation. 13 HIF-1 activation boosted MIF promoter activity and dramatically reduced OGD-induced neuron cell death from a genetic perspective. 48 Finally, in the MCAo model, sex-specific protective benefits were discovered; MIF deletion was detrimental to female mice due to its involvement in regulating inflammation and cell death. 95
When examining particular genetically engineered ischemia models, we see that MIF-KO mice displayed smaller infarct volume at day 7 after 45 min tMCAo, 10 with improved behavioral scores at day 1, 3, and 7 in the same surgical model, as reported by Wang's innovative study. 91 However, another research found that MIF-KO mice had a larger infarct volume 10 hours after 2 hours of tMCAo, with no significant difference at 4 and 22 hours after ischemia. 13 Similar outcomes were seen in MIF-KO mice administered 90 minutes of tMCAo on day 3, with the exception that females had bigger infarcts than WT females but males did not. 95 MIF appears to serve a protective effect at an early stage following prolonged ischemia, but no conclusion has been reached so far. Different observation timepoints, the length of occlusion, or even the origin of genetically engineered animals may account for contradictory MIF function results in ischemic stroke. Further research is required to evaluate the potential bias caused by the model discrepancies noted previously.
The double-faced role of MIF in hemorrhagic stroke
Blood MIF level was reported to be an ideal predictor of delayed cerebral ischemia (DCI) after acute stage of aneurysmal subarachnoid hemorrhage (aSAH), as reported by a single center, prospective, observational cohort study which enrolled 201 patients. 96 This positive correlation between serum MIF concentration and DCI after aSAH was postulated due to the inflammatory nature of MIF, which can exacerbate brain injury and stimulate the immune response during subarachnoid hemorrhage. 96 The difference between the two investigations was that the MIF concentration was measured in either cerebrospinal fluid (CSF) or normal pressure hydrocephalus. 97 These results were comparable to those of an earlier clinical trial using a small sample size. In the study, the MIF concentration in CSF was also identified as a biomarker for aSAH patients who suffered cerebral vasospasm (CV) and DCI sequelae. To obtain more convincing results, a multicenter study with a larger sample size of patients is still required. In patients with intracerebral hemorrhage (ICH), serum MIF concentration rose immediately after disease onset and was strongly correlated with plasma C-reactive protein level, hematoma size, and NIHSS score. 15 Furthermore, it accurately reflected injury severity and can be used as an independent predictor of 6-month unfavorable outcome after acute ICH. 98
Interestingly, compared to clinical data, injecting 2 days before or 2 hours after bacterial collagenase induced experimental intracerebral hemorrhage, MIF alleviated ICH injury by inhibiting microglial activation and macrophage infiltration, resulting in lower reactive oxygen species (ROS) production and neuronal degeneration.14,99
Evidence of MIF functions on traumatic brain injury
Possessing a similar predictive function as stroke, severe blunt trauma patients were also found had higher serum MIF concentrations and related to the clinical outcome. 100 Data from an observational study enrolled 116 severe TBI patients revealed that serum MIF concentrations were also a potential biomarker for identifying cerebrocardiac syndrome (CCS). 101 Another study involving 108 severe TBI found a correlation between the serum MIF level and the severity of TBI and clinical outcomes. 102 In sever blunt trauma patients, Chuang et al.
Wang et al. found that the total expression of MIF remained unchanged in a controlled cortical impact (CCI) induced TBI model. 54 However, MIF entered the nucleus of damaged neurons from the cytosol, causing neurodegeneration with a worsened neurological outcome in a poly ADP-ribose polymerase-1 (PARP-1) dependent manner. 54 Significantly, although MIF can promote neurodegeneration with the participation of CD74, 103 the treatment of MIF antagonist ISO1 failed to prevent neurodegeneration in the peri-injury cortex in a fluid percussion injury (FPI) model of TBI, despite the astrocyte suppression property of MIF during this process. 60
MIF and neurodegenerative diseases
Pleiotropic effects of MIF on Alzheimer’s disease
MIF levels in cerebral spinal fluid and resident brain cells were found to be increased in AD patients and could serve as a biomarker. 104 When compared 52 cognitively healthy volunteers to 97 cognitively impaired or mild dementia patients, the latter had higher MIF levels in CSF, which were significantly correlated with tau and phosphorylated tau protein level. 105 Similarly, another study involving 31 patients with AD, 28 patients with mild cognitive impairment (MCI) and 19 control participants showed that MIF concentrations in CSF were increased in both the AD and MCI groups, with a link between MIF and TNF-α levels in the AD group. 106
MIF was proved to be involved in neuroinflammation and cell toxicity in animal models of AD and in vitro studies.107,108 Pharmacological MIF inhibition or knockout prevented tau protein hyperphosphorylation in C57BL/6 mice, along with better memory improvement in the same streptozotocin induced sporadic AD model.107,109 After treatment with streptozotocin, extracellular MIF levels were considerably increased in cultured astrocytes and microglia, and the MIF antagonist ISO-1 decreased cytokine production in vitro. 107
In the Aβ1-40-stimulated PC12 cell line AD model, treatment with diabetes pathogenic compounds advanced glycation end products (AGEs) led to an increase in MIF expression and a decrease in PC12 cell viability. 110 Consequently, the use of ISO-1 reversed AGEs induced cell deactivation and reduced inflammatory mediators such as IL-1β and TNF-α. 110 In the early stage of AD brains, before the formation of AGEs, fluorescent phenylboronate gel electrophoresis 111 identified glucose-modified MIF that lacked the ability to stimulate enzymes and glial cells, suggesting that MIF participates in the entire progression of high glucose-induced AD-like changes. Using mass spectrometry-based imaging, Carlred et al. confirmed a high degree of co-localization of MIF and activated microglia to amyloid- (A) deposits in the hippocampal region, suggesting that MIF-associated neuroinflammation and glial cell reactivity played a crucial role in the AD pathology. 112 Notably, MIF deficiency impaired the spatial learning function of APP23 transgenic AD mice, suggesting that MIF can shield neuronal cells against A-induced cytotoxicity. The above evidence provides a more complete view of MIF in various AD models. 113
The multifaced role of MIF on Parkinson’s disease
Prior to the year of 2000, the function of MIF on allogeneic fetal mesencephalic dopaminergic grafts was studied using the 6-hydroxydopamine rat model of PD. 114 The intracerebral administration of MIF decreased the activities of microglia and macrophages but didn’t improve the function and survival of grafts. 114 In 2011, Cheng et al. compared 92 PD patients with 87 matched controls and revealed a significant elevation of serum MIF levels in PD patients, proposing MIF levels as a diagnostic biomarker for PD. 115 All clinical studies mentioned in this review were summarized in Table 1.
Table 1.
Clinical investigations of the relationship between MIF and brain injury.
| Disease | Publishing year | Study type and subjects | Clinical samples collection methods | Major findings |
|---|---|---|---|---|
| Ischemic stroke | Liu et al. 2018 64 | Prospective observational study, single center, 39 patients and 14 healthy donors | Blood samples were collected from ischemic stroke patients tPA injection | Plasma MIF level was markedly elevated in the stroke patients |
| Wang et al. 2009 65 | Observational, single center, 102 patients and 57 controls | Not mentioned | The MIF protein level, mRNA levels in PBMCs were significantly higher in the patients than in the controls | |
| Wang et al. 2019 66 | Prospective cohort study with a follow-up for a median of 12 months, single center, 469 patients with ischemic stroke and 423 completed follow-ups | Blood samples from some patients (N = 33) were collected on 12 h, 24 h, 48 h, 72 h, and 96 h after admission for MIF test | Serum levels of MIF increased with increasing severity of stroke and associated with increased risk of stroke recurrence in the next 12 months | |
| Xu et al. 2018 67 | Prospective cohort study with a follow-up of 3 months, single center, 333 patients and 100 healthy volunteers with a three-month follow-up | Fasting blood was collected at 6:00 am on the morning after the admission | Plasma MIF levels were highly associated with infarct volumes and NIHSS score, elevated plasma levels of MIF at admission were associated with increased risk of PSD in the next three months | |
| Wang et al. 2019 68 | Cross-sectional study, single center, 289 patients | Serum was collected under fasting at 8:00 on the first morning of admission and at 12, 24, 48 and 72 h after admission for MIF test | High serum MIF level of stroke patients may be an independent predictor for the moderate to high clinical severity and poor early outcome | |
| Yang et al. 2017 69 | Prospective cohort study, single center, 20 stroke subjects and 14 controls | Peripheral blood samples were collected from healthy control subjects and ischemic stroke subjects at the time of admission | Plasma levels of MIF and the number of CD74-expressing PBMCs were significantly increased in subjects with ischemic stroke at admission, CD74 expression was correlated with stroke severity | |
| Hemorrhagic stroke | Yang et al. 2020 74 | Prospective cohort study with 3 month follow-up, single center, 201 aSAH patients | Fasting serum samples were collected at 6:00 am on the first morning after the admission | Higher level of serum MIF was an independent predictor of DCI in aSAH |
| Kwan et al. 2019 75 | Prospective cohort study, single center, 25 patients in the aSAH group and 9 in the normal pressure hydrocephalus group | CSF was collected from aSAH patients every other day for 13 days from the time of admission up to 72 hours and from NPH patients for 3 days until the lumbar drain was discontinued | CSF MIF concentration in patients with aSAH was significantly higher than those with normal pressure hydrocephalus, CSF concentrations of MIF are correlated with CV and DCI | |
| Lin et al. 2017 76 | Prospective observational study with a follow-up for 6 months, single center, 120 consecutive ICH patients and 120 healthy controls | Blood samples were obtained at admission for the patients | MIF in serum was identified as an independent predictor for 6-month overall survival and 6-month unfavorable outcome after acute ICH | |
| Traumatic brain injury | Chuang et al. 2004 78 | Prospective observational study, single center, 54 severe blunt trauma patients and 44 patients with minor injuries as controls | Peripheral venous blood samples were obtained in the emergency department <4 hrs postinjury (day 1) and the surgical intensive care unit 24 hrs later (day 2) | Serum MIF concentration was higher in severe blunt trauma and correlated with SIRS |
| Dai et al. 2021 79 | prospective observational study, single center, 116 severe TBI patients and 116 healthy controls | Blood samples were collected from controls at study entrance and those of patients were obtained at their arrival to emergency department | Increased serum MIF concentration is independently related to degree of neuroinflammation, clinical severity and risk of cerebrocardiac syndrome in severe TBI patients | |
| Yang et al. 2017 80 | Prospective observational study with a follow-up for 6 months, single center, 108 severe TBI patients and 108 controls | Venous blood was drawn from patients on admission and from controls at study entry | Serum MIF concentrations had close relation to white blood cell count and serum CRP, IL-6 and TNF-α, and it was an independent predictor for long-term prognosis and in-hospital major adverse events |
MIF: macrophage migration inhibitory factor; tPA: tissue plasminogen activator; PBMCs: Peripheral blood mononuclear cells; NIHSS: National Institute of Health stroke scale; PSD: post-stroke depression; aSAH: aneurysmal subarachnoid hemorrhage; DCI: delayed cerebral ischemia; CV: cerebral vasospasm; DCI: delayed cerebral ischemia; CSF: colony stimulating factor; ICH: intracranial hemorrhage; TBI: traumatic brain injury; CRP: C-reactive protein; SIRS: systemic inflammatory response syndrome.
In Atg5 conditional knockout mice, the deficiency of microglial autophagy led to an increase of MIF in a NLRP3 inflammasome, causing neuroinflammation and PD-like symptoms. 66 The MAPKs and NF-κB pathways were implicated in the MIF related LPS-induced PD mouse model. 65 In co-culturing BV-2 microglia and HT22 neuroblastoma cells, the use of a MIF small-molecule inhibitor Z-312 attenuated LPS-induced neurotoxicity with decreased proinflammatory factors; this protective effect of Z-312 was also demonstrated in vivo by a decrease in LPS-induced dopaminergic neuronal loss. 65 For the α-synuclein (α-syn) preformed fibril (PFF) mouse model of sporadic PD, it was recently revealed that genetic and pharmacological inhibition of MIF nuclease activity can prevent neurodegeneration by decreasing PARP-1 activity. 116
Compared with the negative influence on PD pathology above, MIF’s expression was found to be positively correlated with IL-10 and to inhibit apoptosis in SH-SY5Y PD cells via decreasing the concentration of cleaved-PARP. Moreover, MIF enhanced autophagosome production in the SH-SY5Y PD cell model, which may be advantageous for neurodegenerative diseases such as PD. 117 Another neuroprotective effect of MIF was the ability to protect amino acid decarboxylase (AADC)-expressing Chinese hamster ovary (CHO) cells treated with MIF from intracellular but not extracellular dopamine toxicity. 118 Figure 2 shows how MIF interact with AD and PD.
Figure 2.
The double-sided effects of MIF on neurodegenerative disease models. MIF exerts protective effects by inhibiting Aβ-induced cytotoxicity, increasing anti-inflammatory cytokine IL-10 release, promoting autophagosome formation, and reducing intracellular dopamine toxicity in AD and PD models. In terms of the detrimental side, MIF is correlated to tau protein hyperphosphorylation, inflammatory cytokines production, microglial activation, and PARP-1 stimulation. Aβ: amyloid β-protein; AD: Alzheimer’s disease; PD: Parkinsons; PARP-1: poly ADP-ribose polymerase-1 (PARP-1); DA neuron: dopaminergic neuron. The figure were created with BioRender.com.
As previously stated, MIF is a diverse and contentious cytokine in both physiology and pathology. As illness progresses, MIF may display distinct roles in different cell contexts; hence, the impact of varied dosages of MIF on disease progression should not be overlooked.
Concluding remarks
Due to its proinflammatory characteristics, MIF was previously assumed to be deleterious in brain disorders. Recent research has demonstrated that MIF modulation effects are diverse, and that MIF may possibly have organ protective properties in brain damage and neurodegenerative disorders. However, the advantages of MIF in neuroinflammatory diseases were mainly found in basic research, and the specific type of disease model may also influence the explanation of the results. As the numerous activities of MIF in the development of brain damage and neurodegenerative disorders are elucidated, MIF has emerged as a novel and appealing therapeutic target. In MIF-based pharmacological methods, anti-MIF monoclonal antibody, antibodies directed against MIF receptors, and small-molecule inhibitors have been created.119,120 However, there are still limits to these pharmaceutical techniques. Synthesis inhibitors, for instance, may target an array of downstream pathways and lack selectivity. Therefore, MIF must be carefully targeted based on its varied functions.
In this review, we highlight the novel findings of MIF mechanistic insights in immune cells participating in acute and chronic brain disorders, which may shed light on MIF as a prospective therapeutic target in the future.
Acknowledgements
Thank the authors of the references.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: P L. is supported by the National Natural Science Foundation of China (NSFC, 81722017, 91957111, 81971096, 82061130224), New Frontier Technology Joint Research (SHDC12019102) and Ward Building Project for Demonstration and Research sponsored by Shanghai Shenkang Hospital Development Center, Shanghai Municipal Education Commission-Gaofeng Clinical Medical Grant Support (20181805), Shuguang Program supported by Shanghai Education Development Foundation and Shanghai Municipal Education Commission (20SG17), Shanghai Outstanding Academic Leaders’ Program from Shanghai Municipal Science and Technology Committee (20XD1422400), Newton Advanced Fellowship grant provided by the UK Academy of Medical Sciences (NAF\R11\1010), and Renji Clinical Research Funding (PYII20-03) the Innovative Research Team of High-level Local Universities in Shanghai (SHSMU-ZLCX20211602). W.Y. is supported by the Shanghai Pudong New Area Municipal Commission of Health and Family Planning Funding (PWZxq2017-06). Shanghai Municipal Key Clinical Specialty (shslczdzk03601 to Weifeng Yu.), Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation (20DZ2254200). W.X. is supported by the National Natural Science Foundation of China (NSFC, 81901985).
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
ORCID iDs
Yuxuan Lin https://orcid.org/0000-0002-9075-3676
Peiying Li https://orcid.org/0000-0002-5721-9914
References
- 1.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] [PMC free article] [PubMed] [Google Scholar]
- 2.Bach JP, Deuster O, Balzer-Geldsetzer M, et al. The role of macrophage inhibitory factor in tumorigenesis and central nervous system tumors. Cancer 2009; 115: 2031–2040. [DOI] [PubMed] [Google Scholar]
- 3.Nishihira J, Koyama Y, Mizue Y. Identification of macrophage migration inhibitory factor (MIF) in human vascular endothelial cells and its induction by lipopolysaccharide. Cytokine 1998; 10: 199–205. [DOI] [PubMed] [Google Scholar]
- 4.Pan JH, Sukhova GK, Yang JT, et al. Macrophage migration inhibitory factor deficiency impairs atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 2004; 109: 3149–3153. [DOI] [PubMed] [Google Scholar]
- 5.Ogata A, Nishihira J, Suzuki T, et al. Identification of macrophage migration inhibitory factor mRNA expression in neural cells of the rat brain by in situ hybridization. Neurosci Lett 1998; 246: 173–177. [DOI] [PubMed] [Google Scholar]
- 6.Kudrin A, Ray D. Cunning factor: macrophage migration inhibitory factor as a redox-regulated target. Immunol Cell Biol 2008; 86: 232–238. [DOI] [PubMed] [Google Scholar]
- 7.Bacher M, Meinhardt A, Lan HY, et al. MIF expression in the rat brain: implications for neuronal function. Mol Med 1998; 4: 217–230. [PMC free article] [PubMed] [Google Scholar]
- 8.Stefaniak J, Schiefer J, Miller EJ, et al. Macrophage migration inhibitory factor as a potential predictor for requirement of renal replacement therapy after orthotopic liver transplantation. Liver Transpl 2015; 21: 662–669. [DOI] [PubMed] [Google Scholar]
- 9.Noels H, Bernhagen J, Weber C. Macrophage migration inhibitory factor: a noncanonical chemokine important in atherosclerosis. Trends Cardiovasc Med 2009; 19: 76–86. [DOI] [PubMed] [Google Scholar]
- 10.Inácio AR, Ruscher K, Leng L, et al. Macrophage migration inhibitory factor promotes cell death and aggravates neurologic deficits after experimental stroke. J Cereb Blood Flow Metab 2011; 31: 1093–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Li Q, Li Y, Zhang D, et al. Downregulation of microRNA‑451 improves cell migration, invasion and tube formation in hypoxia‑treated HUVECs by targeting MIF. Mol Med Rep 2019; 20: 1167–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chang MC, Park CR, Rhie SH, et al. Early treadmill exercise increases macrophage migration inhibitory factor expression after cerebral ischemia/reperfusion. Neural Regen Res 2019; 14: 1230–1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang S, Zis O, Ly PT, et al. Down-regulation of MIF by NFκB under hypoxia accelerated neuronal loss during stroke. FASEB J 2014; 28: 4394–4407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang J, Tsirka SE. Tuftsin fragment 1-3 is beneficial when delivered after the induction of intracerebral hemorrhage. Stroke 2005; 36: 613–618. [DOI] [PubMed] [Google Scholar]
- 15.Bernhagen J, Mitchell RA, Calandra T, et al. Purification, bioactivity, and secondary structure analysis of mouse and human macrophage migration inhibitory factor (MIF). Biochemistry 1994; 33: 14144–14155. [DOI] [PubMed] [Google Scholar]
- 16.Baugh JA, Chitnis S, Donnelly SC, et al. A functional promoter polymorphism in the macrophage migration inhibitory factor (MIF) gene associated with disease severity in rheumatoid arthritis. Genes Immun 2002; 3: 170–176. [DOI] [PubMed] [Google Scholar]
- 17.Günther S, Fagone P, Jalce G, et al. Role of MIF and D-DT in immune-inflammatory, autoimmune, and chronic respiratory diseases: from pathogenic factors to therapeutic targets. Drug Discov Today 2019; 24: 428–439. [DOI] [PubMed] [Google Scholar]
- 18.Arizza V, Bonura A, La Paglia L, et al. Transcriptional and in silico analyses of MIF cytokine and TLR signalling interplay in the LPS inflammatory response of Ciona robusta. Sci Rep 2020; 10: 11339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rice EK, Tesch GH, Cao Z, et al. Induction of MIF synthesis and secretion by tubular epithelial cells: a novel action of angiotensin II. Kidney Int 2003; 63: 1265–1275. [DOI] [PubMed] [Google Scholar]
- 20.Leng L, Metz CN, Fang Y, et al. MIF signal transduction initiated by binding to CD74. J Exp Med 2003; 197: 1467–1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Alampour-Rajabi SE, Bounkari O, Rot A, et al. MIF interacts with CXCR7 to promote receptor internalization, ERK1/2 and ZAP-70 signaling, and lymphocyte chemotaxis. FASEB J 2015; 29: 4497–4511. [DOI] [PubMed] [Google Scholar]
- 22.Bernhagen J, Krohn R, Lue H, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 2007; 13: 587–596. [DOI] [PubMed] [Google Scholar]
- 23.Gore Y, Starlets D, Maharshak N, et al. Macrophage migration inhibitory factor induces B cell survival by activation of a CD74-CD44 receptor complex. J Biol Chem 2008; 283: 2784–2792. [DOI] [PubMed] [Google Scholar]
- 24.Lue H, Thiele M, Franz J, et al. Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene 2007; 26: 5046–5059. [DOI] [PubMed] [Google Scholar]
- 25.Shi X, Leng L, Wang T, et al. CD44 is the signaling component of the macrophage migration inhibitory factor-CD74 receptor complex. Immunity 2006; 25: 595–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Simi A, Ingelman-Sundberg M, Tindberg N. Neuroprotective agent chlomethiazole attenuates c-fos, c-jun, and AP-1 activation through inhibition of p38 MAP kinase. J Cereb Blood Flow Metab 2000; 20: 1077–1088. [DOI] [PubMed] [Google Scholar]
- 27.Wen Y, Cai W, Yang J, et al. Targeting macrophage migration inhibitory factor in acute pancreatitis and pancreatic cancer. Front Pharmacol 2021; 12: 638950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bemis L, Chan DA, Finkielstein CV, et al. Distinct aerobic and hypoxic mechanisms of HIF-alpha regulation by CSN5. Genes Dev 2004; 18: 739–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Décaillot FM, Kazmi MA, Lin Y, et al. CXCR7/CXCR4 heterodimer constitutively recruits beta-arrestin to enhance cell migration. J Biol Chem 2011; 286: 32188–32197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liu ZY, Song ZW, Guo SW, et al. CXCL12/CXCR4 signaling contributes to neuropathic pain via Central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neurosci Ther 2019; 25: 922–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Saha A, Ahn S, Blando J, et al. Proinflammatory CXCL12-CXCR4/CXCR7 signaling axis drives Myc-induced prostate cancer in obese mice. Cancer Res 2017; 77: 5158–5168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Huang M, Wan Y, Mao L, et al. Inhibiting the migration of M1 microglia at hyperacute period could improve outcome of tMCAO rats. CNS Neurosci Ther 2017; 23: 222–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rafiei S, Gui B, Wu J, et al. Targeting the MIF/CXCR7/AKT signaling pathway in castration-resistant prostate cancer. Mol Cancer Res 2019; 17: 263–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Meiklejohn BH, Coley S. Pressor and catecholamine response to nasal intubation of the trachea. Br J Anaesth 1989; 63: 283–286. [DOI] [PubMed] [Google Scholar]
- 35.West PW, Parker LC, Ward JR, et al. Differential and cell-type specific regulation of responses to toll-like receptor agonists by ISO-1. Immunology 2008; 125: 101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kurita N, Yamashiro K, Kuroki T, et al. Metabolic endotoxemia promotes neuroinflammation after focal cerebral ischemia. J Cereb Blood Flow Metab 2020; 40: 2505–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shin MS, Kang Y, Wahl ER, et al. Macrophage migration inhibitory factor regulates U1 small nuclear RNP immune complex-mediated activation of the NLRP3 inflammasome. Arthritis Rheumatol 2019; 71: 109–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lang T, Lee JPW, Elgass K, et al. Macrophage migration inhibitory factor is required for NLRP3 inflammasome activation. Nat Commun 2018; 9: 2223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Baugh JA, Gantier M, Li L, et al. Dual regulation of macrophage migration inhibitory factor (MIF) expression in hypoxia by CREB and HIF-1. Biochem Biophys Res Commun 2006; 347: 895–903. [DOI] [PubMed] [Google Scholar]
- 40.Morand EF, Leech M, Bernhagen J. MIF: a new cytokine link between rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov 2006; 5: 399–410. [DOI] [PubMed] [Google Scholar]
- 41.Aeberli D, Leech M, Morand EF. Macrophage migration inhibitory factor and glucocorticoid sensitivity. Rheumatology (Oxford) 2006; 45: 937–943. [DOI] [PubMed] [Google Scholar]
- 42.Rogove AD, Tsirka SE. Neurotoxic responses by microglia elicited by excitotoxic injury in the mouse hippocampus. Curr Biol 1998; 8: 19–25. [DOI] [PubMed] [Google Scholar]
- 43.Matsuura T, Sun C, Leng L, et al. Macrophage migration inhibitory factor increases neuronal delayed rectifier K+ current. J Neurophysiol 2006; 95: 1042–1048. [DOI] [PubMed] [Google Scholar]
- 44.Busche S, Gallinat S, Fleegal MA, et al. Novel role of macrophage migration inhibitory factor in angiotensin II regulation of neuromodulation in rat brain. Endocrinology 2001; 142: 4623–4630. [DOI] [PubMed] [Google Scholar]
- 45.Sun C, Li H, Leng L, et al. Macrophage migration inhibitory factor: an intracellular inhibitor of angiotensin II-induced increases in neuronal activity. J Neurosci 2004; 24: 9944–9952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Matsuura T, Harrison RA, Westwell AD, et al. Basal and angiotensin II-inhibited neuronal delayed-rectifier K+ current are regulated by thioredoxin. Am J Physiol Cell Physiol 2007; 293: C211–217. [DOI] [PubMed] [Google Scholar]
- 47.Sun C, Li H, Gao Y, et al. Lack of macrophage migration inhibitory factor regulation is linked to the increased chronotropic action of angiotensin II in SHR neurons. Hypertension 2007; 49: 528–534. [DOI] [PubMed] [Google Scholar]
- 48.Zis O, Zhang S, Dorovini-Zis K, et al. Hypoxia signaling regulates macrophage migration inhibitory factor (MIF) expression in stroke. Mol Neurobiol 2015; 51: 155–167. [DOI] [PubMed] [Google Scholar]
- 49.Ohta S, Misawa A, Fukaya R, et al. Macrophage migration inhibitory factor (MIF) promotes cell survival and proliferation of neural stem/progenitor cells. J Cell Sci 2012; 125: 3210–3220. [DOI] [PubMed] [Google Scholar]
- 50.Bae SH, Yoo MR, Kim YY, et al. Brain-derived neurotrophic factor mediates macrophage migration inhibitory factor to protect neurons against oxygen-glucose deprivation. Neural Regen Res 2020; 15: 1483–1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chai X, Zhang W, Li L, et al. Profile of MIF in developing hippocampus: association with cell proliferation and neurite outgrowth. Front Mol Neurosci 2020; 13: 147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bancroft E, Srinivasan R, Shapiro LA. Macrophage migration inhibitory factor alters functional properties of CA1 hippocampal neurons in mouse brain slices. Int J Mol Sci 2019; 21: 276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhao Y, Wei X, Li W, et al. Inhibition of macrophage migration inhibitory factor protects against inflammation through a toll-like receptor-related pathway after diffuse axonal injury in rats. Biomed Res Int 2020; 2020: 5946205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ruan Z, Lu Q, Wang JE, et al. MIF promotes neurodegeneration and cell death via its nuclease activity following traumatic brain injury. Cell Mol Life Sci 2021; 79: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Balasingam V, Yong VW. Attenuation of astroglial reactivity by interleukin-10. J Neurosci 1996; 16: 2945–2955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bacher M, Weihe E, Dietzschold B, et al. Borna disease virus-induced accumulation of macrophage migration inhibitory factor in rat brain astrocytes is associated with inhibition of macrophage infiltration. Glia 2002; 37: 291–306. [PubMed] [Google Scholar]
- 57.Okazaki S, Boku S, Otsuka I, et al. Clozapine increases macrophage migration inhibitory factor (MIF) expression via increasing histone acetylation of MIF promoter in astrocytes. J Psychiatr Res 2021; 135: 237–242. [DOI] [PubMed] [Google Scholar]
- 58.Di Donato I, Bianchi S, Gallus GN, et al. Heterozygous mutations of HTRA1 gene in patients with familial cerebral small vessel disease. CNS Neurosci Ther 2017; 23: 759–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Fex Svenningsen Å, Löring S, Sørensen AL, et al. Macrophage migration inhibitory factor (MIF) modulates trophic signaling through interaction with serine protease HTRA1. Cell Mol Life Sci 2017; 74: 4561–4572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Newell-Rogers MK, Rogers SK, Tobin RP, et al. Antagonism of macrophage migration inhibitory factory (MIF) after traumatic brain injury ameliorates astrocytosis and peripheral lymphocyte activation and expansion. Int J Mol Sci 2020; 21: 7448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Koda M, Nishio Y, Hashimoto M, et al. Up-regulation of macrophage migration-inhibitory factor expression after compression-induced spinal cord injury in rats. Acta Neuropathol 2004; 108: 31–36. [DOI] [PubMed] [Google Scholar]
- 62.Wang F, Shen X, Guo X, et al. Spinal macrophage migration inhibitory factor contributes to the pathogenesis of inflammatory hyperalgesia in rats. Pain 2010; 148: 275–283. [DOI] [PubMed] [Google Scholar]
- 63.Wang F, Wu H, Xu S, et al. Macrophage migration inhibitory factor activates cyclooxygenase 2-prostaglandin E2 in cultured spinal microglia. Neurosci Res 2011; 71: 210–218. [DOI] [PubMed] [Google Scholar]
- 64.Zhang Y, Gu R, Jia J, et al. Inhibition of macrophage migration inhibitory factor (MIF) tautomerase activity suppresses microglia-mediated inflammatory responses. Clin Exp Pharmacol Physiol 2016; 43: 1134–1144. [DOI] [PubMed] [Google Scholar]
- 65.Zheng LT, Chen J, Zhang L, et al. Inhibition of neuroinflammation by MIF inhibitor 3-({[4-(4-methoxyphenyl)-6-methyl-2-pyrimidinyl]thio}1methyl)benzoic acid (Z-312). Int Immunopharmacol 2021; 98: 107868. [DOI] [PubMed] [Google Scholar]
- 66.Cheng J, Liao Y, Dong Y, et al. Microglial autophagy defect causes Parkinson disease-like symptoms by accelerating inflammasome activation in mice. Autophagy 2020; 16: 2193–2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ghoochani A, Schwarz MA, Yakubov E, et al. MIF-CD74 signaling impedes microglial M1 polarization and facilitates brain tumorigenesis. Oncogene 2016; 35: 6246–6261. [DOI] [PubMed] [Google Scholar]
- 68.Wang J, Lin J, Kaiser U, et al. Absence of macrophage migration inhibitory factor reduces proliferative retinopathy in a mouse model. Acta Diabetol 2017; 54: 383–392. [DOI] [PubMed] [Google Scholar]
- 69.Zernecke A, Bernhagen J, Weber C. Macrophage migration inhibitory factor in cardiovascular disease. Circulation 2008; 117: 1594–1602. [DOI] [PubMed] [Google Scholar]
- 70.Le Hiress M, Tu L, Ricard N, et al. Proinflammatory signature of the dysfunctional endothelium in pulmonary hypertension. Role of the macrophage migration inhibitory factor/CD74 complex. Am J Respir Crit Care Med 2015; 192: 983–997. [DOI] [PubMed] [Google Scholar]
- 71.Li Q, He Q, Baral S, et al. MicroRNA-493 regulates angiogenesis in a rat model of ischemic stroke by targeting MIF. FEBS J 2016; 283: 1720–1733. [DOI] [PubMed] [Google Scholar]
- 72.Liu YC, Tsai YH, Tang SC, et al. Cytokine MIF enhances blood-brain barrier permeability: impact for therapy in ischemic stroke. Sci Rep 2018; 8: 743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.B, Garcia A, Siu E, Du X, et al. Suppression of plasmodium MIF-CD74 signaling protects against severe malaria. FASEB J 2021; 35: e21997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kaplan L, Chow BW, Gu C. Neuronal regulation of the blood-brain barrier and neurovascular coupling. Nat Rev Neurosci 2020; 21: 416–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Vazquez-Liebanas E, Nahar K, Bertuzzi G, et al. Adult-induced genetic ablation distinguishes PDGFB roles in blood-brain barrier maintenance and development. J Cereb Blood Flow Metab 2022; 42: 264–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Stark K, Eckart A, Haidari S, et al. Capillary and arteriolar pericytes attract innate leukocytes exiting through venules and ‘instruct' them with pattern-recognition and motility programs. Nat Immunol 2013; 14: 41–51. [DOI] [PubMed] [Google Scholar]
- 77.Murfee WL, Skalak TC, Peirce SM. Differential arterial/venous expression of NG2 proteoglycan in perivascular cells along microvessels: identifying a venule-specific phenotype. Microcirculation 2005; 12: 151–160. [DOI] [PubMed] [Google Scholar]
- 78.Pellowe AS, Sauler M, Hou Y, et al. Endothelial cell-secreted MIF reduces pericyte contractility and enhances neutrophil extravasation. FASEB J 2019; 33: 2171–2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fu H, Luo F, Yang L, et al. Hypoxia stimulates the expression of macrophage migration inhibitory factor in human vascular smooth muscle cells via HIF-1alpha dependent pathway. BMC Cell Biol 2010; 11: 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Fan Y, Zhang J, Chen CY, et al. Macrophage migration inhibitory factor triggers vascular smooth muscle cell dedifferentiation by a p68-serum response factor axis. Cardiovasc Res 2017; 113: 519–530. [DOI] [PubMed] [Google Scholar]
- 81.Huang H, Chen D, Pu J, et al. The small molecule macrophage migration inhibitory factor antagonist MIF098, inhibits pulmonary hypertension associated with murine SLE. Int Immunopharmacol 2019; 76: 105874. [DOI] [PubMed] [Google Scholar]
- 82.Inácio AR, Bucala R, Deierborg T. Lack of macrophage migration inhibitory factor in mice does not affect hallmarks of the inflammatory/immune response during the first week after stroke. J Neuroinflammation 2011; 8: 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Inácio AR, Ruscher K, Wieloch T. Enriched environment downregulates macrophage migration inhibitory factor and increases parvalbumin in the brain following experimental stroke. Neurobiol Dis 2011; 41: 270–278. [DOI] [PubMed] [Google Scholar]
- 84.Chen Y, Wu X, Yu S, et al. Neuroprotection of tanshinone IIA against cerebral ischemia/reperfusion injury through inhibition of macrophage migration inhibitory factor in rats. PLoS One 2012; 7: e40165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wang J, Ye Q, Xu J, et al. DRα1-MOG-35-55 reduces permanent ischemic brain injury. Transl Stroke Res 2017; 8: 284–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang L, Zis O, Ma G, et al. Upregulation of macrophage migration inhibitory factor gene expression in stroke. Stroke 2009; 40: 973–976. [DOI] [PubMed] [Google Scholar]
- 87.Wang G, Li C, Liu Y, et al. Macrophage migration inhibitory factor levels correlate with stroke recurrence in patients with ischemic stroke. Neurotox Res 2019; 36: 1–11. [DOI] [PubMed] [Google Scholar]
- 88.Xu T, Pu S, Ni Y, et al. Elevated plasma macrophage migration inhibitor factor as a risk factor for the development of post-stroke depression in ischemic stroke. J Neuroimmunol 2018; 320: 58–63. [DOI] [PubMed] [Google Scholar]
- 89.Wang C-W, Ma P-J, Wang Y-Y, et al. Serum level of macrophage migration inhibitory factor predicts severity and prognosis in patients with ischemic stroke. Cytokine 2019; 115: 8–12. [DOI] [PubMed] [Google Scholar]
- 90.Yang L, Kong Y, Ren H, et al. Upregulation of CD74 and its potential association with disease severity in subjects with ischemic stroke. Neurochem Int 2017; 107: 148–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang Y, An R, Umanah GK, et al. A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science 2016; 354: aad6872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hu JX, Ma WJ, He LY, et al. Macrophage migration inhibitory factor (MIF) acetylation protects neurons from ischemic injury. Cell Death Dis 2022; 13: 466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kim JA, Kim YY, Lee SH, et al. Neuroprotective effect of macrophage migration inhibitory factor (MIF) in a mouse model of ischemic stroke. Int J Mol Sci 2022; 23: 6975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jung C, Kim MH, Kim YY, et al. Determining the optimal administration conditions under which MIF exerts neuroprotective effects by inducing BDNF expression and inhibiting apoptosis in an in vitro stroke model. Brain Sci 2021; 11: 280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Turtzo LC, Li J, Persky R, et al. Deletion of macrophage migration inhibitory factor worsens stroke outcome in female mice. Neurobiol Dis 2013; 54: 421–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yang X, Peng J, Pang J, et al. The association between serum macrophage migration inhibitory factor and delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Neurotox Res 2020; 37: 397–405. [DOI] [PubMed] [Google Scholar]
- 97.Kwan K, Arapi O, Wagner KE, et al. Cerebrospinal fluid macrophage migration inhibitory factor: a potential predictor of cerebral vasospasm and clinical outcome after aneurysmal subarachnoid hemorrhage. J Neurosurg 2019; 133: 1786–1791. [DOI] [PubMed] [Google Scholar]
- 98.Lin Q, Cai JY, Lu C, et al. Macrophage migration inhibitory factor levels in serum from patients with acute intracerebral hemorrhage: Potential contribution to prognosis. Clin Chim Acta 2017; 472: 58–63. [DOI] [PubMed] [Google Scholar]
- 99.Wang J, Rogove AD, Tsirka AE, et al. Protective role of tuftsin fragment 1-3 in an animal model of intracerebral hemorrhage. Ann Neurol 2003; 54: 655–664. [DOI] [PubMed] [Google Scholar]
- 100.Chuang CC, Hung CJ, Tsai MC, et al. High concentrations of circulating macrophage migration inhibitory factor in patients with severe blunt trauma: is serum macrophage migration inhibitory factor concentration a valuable prognostic factor? Crit Care Med 2004; 32: 734–739. [DOI] [PubMed] [Google Scholar]
- 101.Dai JX, Lin Q, Ba HJ, et al. Utility of serum macrophage migration inhibitory factor as a potential biomarker for detection of cerebrocardiac syndrome following severe traumatic brain injury. Clin Chim Acta 2021; 512: 179–184. [DOI] [PubMed] [Google Scholar]
- 102.Yang DB, Yu WH, Dong XQ, et al. Serum macrophage migration inhibitory factor concentrations correlate with prognosis of traumatic brain injury. Clin Chim Acta 2017; 469: 99–104. [DOI] [PubMed] [Google Scholar]
- 103.Tobin RP, Mukherjee S, Kain JM, et al. Traumatic brain injury causes selective, CD74-dependent peripheral lymphocyte activation that exacerbates neurodegeneration. Acta Neuropathol Commun 2014; 2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bacher M, Deuster O, Aljabari B, et al. The role of macrophage migration inhibitory factor in Alzheimer's disease. Mol Med 2010; 16: 116–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Oikonomidi A, Tautvydaitė D, Gholamrezaee MM, et al. Macrophage migration inhibitory factor is associated with biomarkers of Alzheimer's disease pathology and predicts cognitive decline in mild cognitive impairment and mild dementia. JAD 2017; 60: 273–281. [DOI] [PubMed] [Google Scholar]
- 106.Popp J, Bacher M, Kölsch H, et al. Macrophage migration inhibitory factor in mild cognitive impairment and Alzheimer's disease. J Psychiatr Res 2009; 43: 749–753. [DOI] [PubMed] [Google Scholar]
- 107.Nasiri E, Sankowski R, Dietrich H, et al. Key role of MIF-related neuroinflammation in neurodegeneration and cognitive impairment in Alzheimer's disease. Mol Med 2020; 26: 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liang CJ, Li JH, Zhang Z, et al. Suppression of MIF-induced neuronal apoptosis may underlie the therapeutic effects of effective components of Fufang Danshen in the treatment of Alzheimer's disease. Acta Pharmacol Sin 2018; 39: 1421–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Li SQ, Yu Y, Han JZ, et al. Deficiency of macrophage migration inhibitory factor attenuates tau hyperphosphorylation in mouse models of Alzheimer's disease. J Neuroinflammation 2015; 12: 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yu M, Zang D, Xu Y, et al. Protective effect of ISO‑1 against advanced glycation end product aggravation of PC12 cell injury induced by Aβ1‑40. Mol Med Rep 2019; 20: 2135–2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kassaar O, P, Morais M, Xu S, et al. Macrophage migration inhibitory factor is subjected to glucose modification and oxidation in Alzheimer's disease. Sci Rep 2017; 7: 42874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Carlred L, Michno W, Kaya I, et al. Probing amyloid-β pathology in transgenic Alzheimer's disease (tgArcSwe) mice using MALDI imaging mass spectrometry. J Neurochem 2016; 138: 469–478. [DOI] [PubMed] [Google Scholar]
- 113.Zhang S, Zhao J, Zhang Y, et al. Upregulation of MIF as a defense mechanism and a biomarker of Alzheimer's disease. Alzheimers Res Ther 2019; 11: 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Schwarz SC, Schwarz J, Sautter J, et al. Effects of macrophage migration inhibitory factor and macrophage migration stimulatory factor on function and survival of foetal dopaminergic grafts in the 6-hydroxydopamine rat model of Parkinson's disease. Exp Brain Res 1998; 120: 95–103. [DOI] [PubMed] [Google Scholar]
- 115.Nicoletti A, Fagone P, Donzuso G, et al. Parkinson's disease is associated with increased serum levels of macrophage migration inhibitory factor. Cytokine 2011; 55: 165–167. [DOI] [PubMed] [Google Scholar]
- 116.Park H, Kam TI, Peng H, et al. PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson's disease. Cell 2022; 185: 1943–1959.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Li S, Nie K, Zhang Q, et al. Macrophage migration inhibitory factor mediates neuroprotective effects by regulating inflammation, apoptosis and autophagy in Parkinson's disease. Neuroscience 2019; 416: 50–62. [DOI] [PubMed] [Google Scholar]
- 118.Weingarten P, Zhou QY. Protection of intracellular dopamine cytotoxicity by dopamine disposition and metabolism factors. J Neurochem 2001; 77: 776–785. [DOI] [PubMed] [Google Scholar]
- 119.Sumaiya K, Langford D, Natarajaseenivasan K, et al. Macrophage migration inhibitory factor (MIF): a multifaceted cytokine regulated by genetic and physiological strategies. Pharmacol Ther 2022; 233: 108024. [DOI] [PubMed] [Google Scholar]
- 120.Kang I, Bucala R. The immunobiology of MIF: function, genetics and prospects for precision medicine. Nat Rev Rheumatol 2019; 15: 427–437. [DOI] [PubMed] [Google Scholar]


