1
|
Juárez-Avelar I, Rodríguez T, García-García AP, Rodríguez-Sosa M. Macrophage migration inhibitory factor (MIF): Its role in the genesis and progression of colorectal cancer. IMMUNOTHERAPY IN RESISTANT CANCER: FROM THE LAB BENCH WORK TO ITS CLINICAL PERSPECTIVES 2021:173-193. [DOI: 10.1016/b978-0-12-822028-3.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Cirillo PF, Asojo OA, Khire U, Lee Y, Mootien S, Hegan P, Sutherland AG, Peterson-Roth E, Ledizet M, Koski RA, Anthony KG. Inhibition of Macrophage Migration Inhibitory Factor by a Chimera of Two Allosteric Binders. ACS Med Chem Lett 2020; 11:1843-1847. [PMID: 33062162 DOI: 10.1021/acsmedchemlett.9b00351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/19/2019] [Indexed: 01/05/2023] Open
Abstract
Human Macrophage Migration Inhibitory Factor (MIF) is a trimeric cytokine implicated in a number of inflammatory and autoimmune diseases and cancer. We previously reported that the dye p425 (Chicago Sky Blue), which bound MIF at the interface of two MIF trimers covering the tautomerase and allosteric pockets, revealed a unique strategy to block MIF's pro-inflammatory activities. Structural liabilities, including the large size, precluded p425 as a medicinal chemistry lead for drug development. We report here a rational design strategy linking only the fragment of p425 that binds over the tautomerase pocket to the core of ibudilast, a known MIF allosteric site-specific inhibitor. The chimeric compound, termed L2-4048, was shown by X-ray crystallography to bind at the allosteric and tautomerase sites as anticipated. L2-4048 retained target binding and blocked MIF's tautomerase CD74 receptor binding, and pro-inflammatory activities. Our studies lay the foundation for the design and synthesis of smaller and more drug-like compounds that retain the MIF inhibitory properties of this chimera.
Collapse
Affiliation(s)
- Pier F. Cirillo
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
- Department of Chemistry and Chemical Engineering, University of New Haven, 300 Boston Post Road, West Haven, Connecticut 06516, United States
| | - Oluwatoyin A. Asojo
- Department of Chemistry and Biochemistry, Hampton University, 200 William R. Harvey Way, Hampton, Virginia 23668, United States
| | - Uday Khire
- CheminPharma, LLC, 4 Research Drive, Woodbridge, Connecticut 06525, United States
| | - Yashang Lee
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | - Sara Mootien
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | - Peter Hegan
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | - Alan G. Sutherland
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | | | - Michel Ledizet
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | - Raymond A. Koski
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| | - Karen G. Anthony
- L2 Diagnostics, LLC, 300 George Street, New Haven, Connecticut 06511, United States
| |
Collapse
|
3
|
Li S, Nie K, Zhang Q, Guo M, Qiu Y, Li Y, Gao Y, Wang L. Macrophage Migration Inhibitory Factor Mediates Neuroprotective Effects by Regulating Inflammation, Apoptosis and Autophagy in Parkinson's Disease. Neuroscience 2019; 416:50-62. [DOI: 10.1016/j.neuroscience.2019.05.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/15/2023]
|
4
|
Stošić-Grujičić S, Saksida T, Miljković Đ, Stojanović I. MIF and insulin: Lifetime companions from common genesis to common pathogenesis. Cytokine 2019; 125:154792. [PMID: 31400637 DOI: 10.1016/j.cyto.2019.154792] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/01/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022]
Abstract
Pro-inflammatory nature of macrophage migration inhibitory factor (MIF) has been generally related to the propagation of inflammatory and autoimmune diseases. But this molecule possesses many other peculiar functions, unrelated to the immune system, among which is its supportive role in the post-translational modifications of insulin. In this way MIF enables proper insulin conformation within the pancreatic beta cell and its full activity. The inherent or acquired changes in MIF expression might therefore lead to different insulin processing and initiation of autoimmunity. The relation between MIF and insulin does not stop at this point; these two molecules continue to interact during pathological states characterized by inflammation and insulin resistance. In this context, MIF indirectly and negatively influences insulin action by boosting inflammatory environment and disabling target cells to respond to insulin. On the other side, insulin might interfere with MIF action as well, acting as an anti-inflammatory mediator. Therefore, the proper interaction between MIF and insulin is crucial for maintaining homeostasis, while anti-inflammatory therapies based on the systemic MIF blockage may disturb this balance. This review covers MIF-insulin relationship in the physiological and pathological conditions and discusses the approaches for MIF inhibition and their net effect specifically considering possible impact on insulin misfolding and the possible misinterpretation of previous results due to the discovery of MIF functional homolog D-dopachrome tautomerase.
Collapse
Affiliation(s)
- Stanislava Stošić-Grujičić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
5
|
Kok T, Wasiel AA, Cool RH, Melgert BN, Poelarends GJ, Dekker FJ. Small-molecule inhibitors of macrophage migration inhibitory factor (MIF) as an emerging class of therapeutics for immune disorders. Drug Discov Today 2018; 23:1910-1918. [PMID: 29936245 DOI: 10.1016/j.drudis.2018.06.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/18/2018] [Accepted: 06/18/2018] [Indexed: 01/22/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is an important cytokine for which an increasing number of functions is being described in the pathogenesis of inflammation and cancer. Nevertheless, the availability of potent and druglike MIF inhibitors that are well-characterized in relevant disease models remains limited. Development of highly potent and selective small-molecule MIF inhibitors and validation of their use in relevant disease models will advance drug discovery. In this review, we provide an overview of recent advances in the identification of MIF as a pharmacological target in the pathogenesis of inflammatory diseases and cancer. We also give an overview of the current developments in the discovery and design of small-molecule MIF inhibitors and define future aims in this field.
Collapse
Affiliation(s)
- Tjie Kok
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands; Faculty of Biotechnology, University of Surabaya, Jalan Raya Kalirungkut Surabaya, 60292, Indonesia
| | - Anna A Wasiel
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Robbert H Cool
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Barbro N Melgert
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerrit J Poelarends
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
6
|
Trivedi-Parmar V, Jorgensen WL. Advances and Insights for Small Molecule Inhibition of Macrophage Migration Inhibitory Factor. J Med Chem 2018; 61:8104-8119. [PMID: 29812929 DOI: 10.1021/acs.jmedchem.8b00589] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream regulator of the immune response whose dysregulation is tied to a broad spectrum of inflammatory and proliferative disorders. As its complex signaling pathways and pleiotropic nature have been elucidated, it has become an attractive target for drug discovery. Remarkably, MIF is both a cytokine and an enzyme that functions as a keto-enol tautomerase. Strategies including in silico modeling, virtual screening, high-throughput screening, and screening of anti-inflammatory natural products have led to a large and diverse catalogue of MIF inhibitors as well as some understanding of the structure-activity relationships for compounds binding MIF's tautomerase active site. With possible clinical trials of some MIF inhibitors on the horizon, it is an opportune time to review the literature to seek trends, address inconsistencies, and identify promising new avenues of research.
Collapse
Affiliation(s)
- Vinay Trivedi-Parmar
- Department of Chemistry , Yale University , New Haven , Connecticut 06520-8107 , United States
| | - William L Jorgensen
- Department of Chemistry , Yale University , New Haven , Connecticut 06520-8107 , United States
| |
Collapse
|
7
|
Sparkes A, De Baetselier P, Brys L, Cabrito I, Sterckx YGJ, Schoonooghe S, Muyldermans S, Raes G, Bucala R, Vanlandschoot P, Van Ginderachter JA, Stijlemans B. Novel half-life extended anti-MIF nanobodies protect against endotoxic shock. FASEB J 2018; 32:3411-3422. [PMID: 29401625 DOI: 10.1096/fj.201701189r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sepsis-leading to septic shock-is the leading cause of death in intensive care units. The systemic inflammatory response to infection, which is initiated by activated myeloid cells, plays a key role in the lethal outcome. Macrophage migration inhibitory factor (MIF) is an upstream immunoregulatory mediator, released by myeloid cells, that underlies a common genetic susceptibility to different infections and septic shock. Accordingly, strategies that are aimed at inhibiting the action of MIF have therapeutic potential. Here, we report the isolation and characterization of tailorable, small, affinity-matured nanobodies (Nbs; single-domain antigen-binding fragments derived from camelid heavy-chain Abs) directed against MIF. Of importance, these bioengineered Nbs bind both human and mouse MIFs with nanomolar affinity. NbE5 and NbE10 inhibit key MIF functions that can exacerbate septic shock, such as the tautomerase activity of MIF (by blocking catalytic pocket residues that are critical for MIF's conformation and receptor binding), the TNF-inducing potential, and the ability of MIF to antagonize glucocorticoid action. A lead NbE10, tailored to be a multivalent, half-life extended construct (NbE10-NbAlb8-NbE10), attenuated lethality in murine endotoxemia when administered via single injection, either prophylactically or therapeutically. Hence, Nbs, with their structural and pharmacologic advantages over currently available inhibitors, may be an effective, novel approach to interfere with the action of MIF in septic shock and other conditions of inflammatory end-organ damage.-Sparkes, A., De Baetselier, P., Brys, L., Cabrito, I., Sterckx, Y. G.-J., Schoonooghe, S., Muyldermans, S., Raes, G., Bucala, R., Vanlandschoot, P., Van Ginderachter, J. A., Stijlemans, B. Novel half-life extended anti-MIF nanobodies protect against endotoxic shock.
Collapse
Affiliation(s)
- Amanda Sparkes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Patrick De Baetselier
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Lea Brys
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Inês Cabrito
- Department of Biopharmaceuticals, Pharmaceutical Product Development (PPD) Laboratories, Good Manufacturing Practices (GMP) Laboratory, Athlone, Ireland.,Ablynx NV, Zwijnaarde, Belgium
| | - Yann G-J Sterckx
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium
| | - Steve Schoonooghe
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| | - Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Flanders Institute for Biotechnology (VIB) Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
8
|
Zheng S, Ren X, Han T, Chen Y, Qiu H, Liu W, Hu Y. Fenofibrate attenuates fatty acid-induced islet β-cell dysfunction and apoptosis via inhibiting the NF-κB/MIF dependent inflammatory pathway. Metabolism 2017; 77:23-38. [PMID: 28941594 DOI: 10.1016/j.metabol.2017.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Fatty acid-induced lipotoxicity and macrophage migration inhibitory factor (MIF) affect pancreatic β-cell function, and may promote the development of diabetes mellitus. However, the association of lipotoxicity with MIF and the effect of Fenofibrate on β-cell function remain unknown. METHODS LPL+/- mice and MIN6 cells stimulated with palmitic acid (PA) were utilized as models of lipid metabolism disorders. Factors associated with insulin secretion and apoptosis were assessed in the presence or absence of Fenofibrate. The possible mechanisms of lipotoxicity-induced β-cell dysfunction were also explored. RESULTS Fenofibrate effectively improved lipid accumulation in pancreatic β-cells, increased glucose-stimulated insulin secretion and β-cell mass, and significantly downregulated pro-apoptotic molecules, at the gene and protein levels, both in vivo and in vitro. Additionally, elevated MIF levels in serum from LPL+/- mice and PA-treated MIN6 cells were starkly decreased after Fenofibrate administration. Mechanistic analysis indicated that NF-κB signaling was remarkably triggered, which could further activate MIF transcription. Furthermore, Fenofibrate exerted beneficial effects on fatty acid-induced β-cell dysfunction likely by inhibiting the NF-κB/MIF dependent inflammatory response. CONCLUSIONS Fenofibrate ameliorates lipotoxicity-induced β-cell dysfunction and apoptosis by inhibiting the NF-κB/MIF inflammatory pathway. These findings provide novel insights into the treatment of lipotoxicity-induced metabolic disorders.
Collapse
Affiliation(s)
- Shuang Zheng
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xingxing Ren
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Tingting Han
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yawen Chen
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Huiying Qiu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei Liu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yaomin Hu
- Department of Endocrinology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
9
|
Bloom J, Metz C, Nalawade S, Casabar J, Cheng KF, He M, Sherry B, Coleman T, Forsthuber T, Al-Abed Y. Identification of Iguratimod as an Inhibitor of Macrophage Migration Inhibitory Factor (MIF) with Steroid-sparing Potential. J Biol Chem 2016; 291:26502-26514. [PMID: 27793992 DOI: 10.1074/jbc.m116.743328] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/27/2016] [Indexed: 12/11/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been implicated in a broad range of inflammatory and oncologic diseases. MIF is unique among cytokines in terms of its release profile and inflammatory role, notably as an endogenous counter-regulator of the anti-inflammatory effects of glucocorticoids. In addition, it exhibits a catalytic tautomerase activity amenable to the design of high affinity small molecule inhibitors. Although several classes of these compounds have been identified, biologic characterization of these molecules remains a topic of active investigation. In this study, we used in vitro LPS-driven assays to characterize representative molecules from several classes of MIF inhibitors. We determined that MIF inhibitors exhibit distinct profiles of anti-inflammatory activity, especially with regard to TNFα. We further investigated a molecule with relatively low anti-inflammatory activity, compound T-614 (also known as the anti-rheumatic drug iguratimod), and found that, in addition to exhibiting selective MIF inhibition in vitro and in vivo, iguratimod also has additive effects with glucocorticoids. Furthermore, we found that iguratimod synergizes with glucocorticoids in attenuating experimental autoimmune encephalitis, a model of multiple sclerosis. Our work identifies iguratimod as a valuable new candidate for drug repurposing to MIF-relevant diseases, including multiple sclerosis.
Collapse
Affiliation(s)
- Joshua Bloom
- From the Hofstra-Northwell School of Medicine, Hempstead, New York 11549, .,the Centers for Molecular Innovation
| | - Christine Metz
- From the Hofstra-Northwell School of Medicine, Hempstead, New York 11549.,Biomedical Sciences, and
| | - Saisha Nalawade
- the Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249
| | - Julian Casabar
- the Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249
| | | | | | - Barbara Sherry
- From the Hofstra-Northwell School of Medicine, Hempstead, New York 11549.,Immunology and Inflammation, and
| | - Thomas Coleman
- the Office of Technology Transfer, The Feinstein Institute for Medical Research, Manhasset, New York 11030, and
| | - Thomas Forsthuber
- the Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249
| | - Yousef Al-Abed
- From the Hofstra-Northwell School of Medicine, Hempstead, New York 11549, .,the Centers for Molecular Innovation
| |
Collapse
|
10
|
Cisneros JA, Robertson MJ, Valhondo M, Jorgensen WL. Irregularities in enzyme assays: The case of macrophage migration inhibitory factor. Bioorg Med Chem Lett 2016; 26:2764-2767. [PMID: 27156768 DOI: 10.1016/j.bmcl.2016.04.074] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 04/21/2016] [Accepted: 04/23/2016] [Indexed: 12/17/2022]
Abstract
Inhibitors of human macrophage migration inhibitory factor (MIF) previously reported in the literature have been reexamined by synthesis, assaying for tautomerase activity, and protein crystallography. Substantial inconsistencies between prior and current assay results are noted. They appear to arise from difficulties with the tautomerase substrates, solubility issues, and especially covalent inhibition. Incubation time variation shows that 3, 4, 6, and 9 are covalent or slow-binding inhibitors. Two protein crystal structures are provided; one confirms that the twice-discovered 3 is a covalent inhibitor.
Collapse
Affiliation(s)
- José A Cisneros
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States
| | - Michael J Robertson
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States
| | - Margarita Valhondo
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States
| | - William L Jorgensen
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States.
| |
Collapse
|
11
|
Berberine Induces Cell Apoptosis through Cytochrome C/Apoptotic Protease-Activating Factor 1/Caspase-3 and Apoptosis Inducing Factor Pathway in Mouse Insulinoma Cells. Chin J Integr Med 2015; 25:853-860. [DOI: 10.1007/s11655-015-2280-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2015] [Indexed: 12/20/2022]
|