Elsevier

European Journal of Pharmacology

Volume 740, 5 October 2014, Pages 683-689
European Journal of Pharmacology

Immunopharmacology and inflammation
Novel inhibitors of macrophage migration inhibitory factor prevent cytokine-induced beta cell death

https://doi.org/10.1016/j.ejphar.2014.06.009Get rights and content

Abstract

Macrophage migration inhibitory factor is a multifunctional cytokine involved in the regulation of immune processes and also in apoptosis induction. Elevated MIF expression is detrimental for insulin-producing beta cells and MIF inhibition protected beta cells from several cytotoxic insults such as inflammatory cytokines, high fatty acids or high glucose concentrations. Therefore, the aim of this study was to investigate two newly synthesized small molecule MIF inhibitors (K664-1 and K647-1) and to compare them with previously established effects of the prototypical MIF inhibitor, ISO-1. Our results indicate that K664-1 and K647-1 are 160- and 40-fold more effective in inhibition of MIF׳s tautomerase activity than ISO-1. Also, new inhibitors confer beta cell protection from cytokine-triggered apoptosis at significantly lower concentrations than ISO-1. Although all three MIF inhibitors inhibit caspase 3 activity, K664-1 and K647-1 suppress pro-apoptotic BAX protein expression and up-regulate anti-apoptotic Bcl-2 mRNA. Finally, all three MIF inhibitors operate through blockade of nitric oxide production stimulated by cytokines. In conclusion, two novel MIF inhibitors are more potent than ISO-1 and operate through inhibition of the mitochondria-related apoptotic pathway. We propose that these compounds represent a unique class of anti-MIF antagonists that should be further tested for therapeutic use.

Introduction

Type 1diabetes is an autoimmune disease characterized by a local inflammatory reaction in and around the pancreatic islets, followed by progressive destruction of insulin producing beta cells that leads to hyperglycemia. Until now, a number of pathological stimuli have been identified to induce beta cell apoptosis: reactive oxidative and nitrogen species, perforins and granzymes, Fas ligand and most importantly pro-inflammatory cytokines such as interferon-γ (IFN-γ), interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α) (Padgett et al., 2013). Type 2 diabetes is characterized by insulin resistance in peripheral tissues that manifests as hyperglycemia when beta cells fail to regulate insulin production due to their demise mediated by elevated nutrients and cytokines. All mentioned cytotoxic stimuli that contribute to beta cell loss identified in type 1 and type 2 diabetes converge on mitochondria to lead the beta cell to apoptosis (Szabadkai and Duchen, 2009). Recently, another cytokine, macrophage migration inhibitory factor (MIF) has been identified as an important factor for beta cell survival and MIF inhibition plays a crucial role in blocking a mitochondria-mediated apoptotic process. MIF absence or blockade ensures protection of beta cells from exposure to either pro-inflammatory cytokines (Stojanovic et al., 2012a) or elevated nutrients (palmitic acid or glucose) in vitro (Stojanovic et al., 2012b).

in vivo experiments proved that MIF is crucial for development of type 1 diabetes (Bojunga et al., 2003) and that MIF inhibition by neutralizing antibodies (Cvetkovic et al., 2005) or complete MIF absence (MIF knock-out) ameliorates initiation and/or progression of type 1 diabetes (Stosic-Grujicic et al., 2008). These data indicate that the inhibition of MIF could be exploited as a therapeutic strategy (Stosic-Grujicic et al., 2009, Greven et al., 2010). Having in mind that the usage of neutralizing antibodies for therapy could result in side-effects and is expensive, several pharmacologically designed MIF inhibitors have been synthetized and have shown promising results. One of the best characterized and also most well-known MIF inhibitors is (S,R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1) (reviewed in Al-Abed et al., 2011). It was shown that ISO-1 has beneficial effects in experimental colitis (Shah et al., 2008), experimental allergic neuritis (Nicoletti et al., 2005) and in endotoxemia (Al-Abed et al., 2005). In MIF׳s homotrimeric structure a hydrophobic pocket region formed between each adjacent subunit plays an important role in its biological function, as evidenced by the fact that ISO-1 binding of this region decreases downstream MIF signaling, MIF biological activities and clinical outcomes that are associated with MIF (Al-Abed et al., 2011).

Since we have previously shown that in vivo MIF inhibition by ISO-1 ameliorates streptozotocin induced type 1 diabetes development in C57BL/6 mice (Cvetkovic et al., 2005) and that in vitro ISO-1 protects insulinoma cells or pancreatic islets from pro-inflammatory cytokines (Stojanovic et al., 2008) the aim of this study was to compare the activity of two novel MIF inhibitors with ISO-1 and to investigate, in more detail, the mechanism of their action.

Section snippets

Materials and methods

Unless stated otherwise, all chemicals were from Sigma-Aldrich (St Louis, MO, USA) and all plastics used in the experiments were from Sardstedt (Numbrecht, Germany).

Novel MIF inhibitors K664-1 and K647-1 are potent inhibitors of MIF enzymatic activity

The concentration required to inhibit 50% of the control tautomerase activity (IC-50) was determined by established methods and compared for the two new compounds versus the established MIF inhibitor, ISO-1. The IC-50′s of K647-1 (0.41×0.01 μM) and K664-1 (0.11±0.03 μM) were approximately 40 and 160x more potent than ISO-1 (Table 1).

K664-1 and K647-1 inhibit the recognition of MIF

Interaction of ISO-1 with recombinant MIF in vitro down-regulates immunorecognition of MIF during ELISA (Hegde et al., 2012). Therefore, we first sought to

Discussion

In the present study, we demonstrate that the two newly synthetized compounds, K664-1 and K647-1, are potent inhibitors of MIF tautomerase activity and that they rescue beta cells from cytokine-driven apoptosis. This protection is provided at lower concentrations compared to ISO-1. The new MIF inhibitors likely operate through inhibition of BAX-mediated apoptosis, unlike ISO-1. However, all three inhibitors prevent caspase-3 activation and NO production.

MIF is an important regulator of

Acknowledgments

We would like to thank Melita Vidakovic, PhD, from the Institute for Biological Research “Sinisa Stankovic”, University of Belgrade, Belgrade, Serbia for the kind gift of RINm5f cell culture. This work was supported by Ministry of Education, Science and Technological Development, Republic of Serbia (Project no. 173013).

References (28)

  • I. Cvetkovic et al.

    Critical role of macrophage migration inhibitory factor activity in experimental autoimmune diabetes

    Endocrinology

    (2005)
  • A. Dios et al.

    Inhibition of MIF bioactivity by rational design of pharmacological inhibitors of MIF tautomerase activity

    J. Med. Chem.

    (2002)
  • G. Fingerle-Rowson 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)
  • D. Greven et al.

    Autoimmune diseases: MIF as a therapeutic target

    Expert Opin. Ther. Targets

    (2010)
  • Cited by (11)

    • Macrophage migration inhibitory factor (MIF): Its role in the genesis and progression of colorectal cancer

      2020, Immunotherapy in Resistant Cancer: From the Lab Bench Work to Its Clinical Perspectives
    • MIF and insulin: Lifetime companions from common genesis to common pathogenesis

      2020, Cytokine
      Citation Excerpt :

      This issue will be discussed further in the next section. Intriguingly, both genetic MIF deletion and MIF inhibition with different MIF inhibitors prevented cytokine-induced beta cell death through down-regulation of BAX or caspase 3-related pathways in vitro [61,62]. This means that in the circumstances of MIF overproduction (under the influence of cytokines or palmitic acid), MIF acquires detrimental influence on beta cells.

    • Macrophage Migration Inhibitory Factor Mediates Neuroprotective Effects by Regulating Inflammation, Apoptosis and Autophagy in Parkinson's Disease

      2019, Neuroscience
      Citation Excerpt :

      MIF upregulated and knock-down PD cells model was successful established using lentivirus (P < 0.001; Fig. 2D) and siRNA (P < 0.01; Fig. 2E), respectively, for use in subsequent experiments. According to the above results, ISO-1, an inhibitor of MIF (Vujicic et al., 2014) (Fig. 2F), was added in order to verify the role of MIF in the MPP + SH-SY5Y cell model. As shown in Fig. 2G, the expression of MIF was significantly downregulated in the MPP + SH-SY5Y cell model upon ISO-1 addition (P < 0.001).

    • Small-molecule inhibitors of macrophage migration inhibitory factor (MIF) as an emerging class of therapeutics for immune disorders

      2018, Drug Discovery Today
      Citation Excerpt :

      Another study, reported for MIF inhibitor K664-1 (Table 2) with a pyrimidazole scaffold an IC50 of 0.16 μM in the MIF tautomerase assay [72]. This inhibitor provided protection to β cells from cytokine-triggered apoptosis in a mouse model, which demonstrates its potential for the prevention of diabetes progression [73]. In 2016, compound T-614 (also known as iguratimod) was found to selectively inhibit MIF in vitro and in vivo.

    • Fenofibrate attenuates fatty acid-induced islet β-cell dysfunction and apoptosis via inhibiting the NF-κB/MIF dependent inflammatory pathway

      2017, Metabolism: Clinical and Experimental
      Citation Excerpt :

      Additionally, MIF may act as a pro-apoptotic molecule in β-cells after exposure to any toxic insult. MIF triggers apoptosis via the mitochondria-related apoptotic pathway by decreasing the Bcl-2/Bax ratio, with subsequent cleavage of caspase 3 in pancreatic β-cells [47]. Consistent with these findings, the increased expression levels of Bax and cleaved caspase 3 in both animal and cell models were also observed in our study.

    View all citing articles on Scopus
    1

    Authors equally contributed.

    View full text