1
|
Zhang L, Zhang H, Agborbesong E, Zhou JX, Li X. Phosphorylation of MIF by PIP4K2a is necessary for cilia biogenesis. Cell Death Dis 2023; 14:795. [PMID: 38052787 PMCID: PMC10698143 DOI: 10.1038/s41419-023-06323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023]
Abstract
Primary cilia are microtubule-based organelles that play important roles in development and tissue homeostasis. Macrophage migration inhibitory factor (MIF) has long been recognized as a secreted cytokine in the pathogenesis of various human diseases, including cancer and autosomal dominant polycystic kidney disease (ADPKD). Unlike other cytokines, unique functional characteristics of intracellular MIF have emerged. In this study, we show that MIF is localized and formed a ring like structure at the proximal end of centrioles, where it regulates cilia biogenesis through affecting 1) the recruitment of TTBK2 to basal body and the removal of CP110 from mother centriole, 2) the accumulation of CEP290 at centriolar satellites, and 3) the trafficking of intraflagellar transport (IFT) related proteins. We also show that MIF functions as a novel transcriptional factor to regulate the expression of genes related to ciliogenesis via binding on the promotors of those genes. MIF also binds chromatin and regulates transcription of genes involved in diverse homeostatic signaling pathways. We identify phosphatidylinositol-5-phosphate 4-kinase type 2 alpha (PIP4K2a) as an upstream regulator of MIF, which interacts with and phosphorylates MIF at S91 to increase its interaction with 14-3-3ζ, resulting in its nuclear translocation and transcription regulation. This study suggests that MIF is a key player in cilia biogenesis and a novel transcriptional regulator in homeostasis, which forward our understanding of how MIF is able to carry out several nonoverlapping functions.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hongbing Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
Yoon K. Gastric Cancer: H. pylori and Macrophage Migration Inhibitory Factor. HELICOBACTER PYLORI 2023:321-326. [DOI: 10.1007/978-981-97-0013-4_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Winkler R, Piskor EM, Kosan C. Lessons from Using Genetically Engineered Mouse Models of MYC-Induced Lymphoma. Cells 2022; 12:37. [PMID: 36611833 PMCID: PMC9818924 DOI: 10.3390/cells12010037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022] Open
Abstract
Oncogenic overexpression of MYC leads to the fatal deregulation of signaling pathways, cellular metabolism, and cell growth. MYC rearrangements are found frequently among non-Hodgkin B-cell lymphomas enforcing MYC overexpression. Genetically engineered mouse models (GEMMs) were developed to understand MYC-induced B-cell lymphomagenesis. Here, we highlight the advantages of using Eµ-Myc transgenic mice. We thoroughly compiled the available literature to discuss common challenges when using such mouse models. Furthermore, we give an overview of pathways affected by MYC based on knowledge gained from the use of GEMMs. We identified top regulators of MYC-induced lymphomagenesis, including some candidates that are not pharmacologically targeted yet.
Collapse
Affiliation(s)
| | | | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich Schiller University Jena, 07745 Jena, Germany
| |
Collapse
|
4
|
Thavayogarajah T, Sinitski D, Bounkari OE, Torres-Garcia L, Lewinsky H, Harjung A, Chen HR, Panse J, Vankann L, Shachar I, Bernhagen J, Koschmieder S. CXCR4 and CD74 together enhance cell survival in response to macrophage migration-inhibitory factor in chronic lymphocytic leukemia. Exp Hematol 2022; 115:30-43. [PMID: 36096455 DOI: 10.1016/j.exphem.2022.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 11/04/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of small, mature CD5+ B lymphocytes in the blood, marrow, and lymphoid organs. Cell survival depends on interaction with the leukemic microenvironment. However, the mechanisms controlling CLL cell survival are still incompletely understood. Macrophage migration-inhibitory factor (MIF), a pro-inflammatory and immunoregulatory chemokine-like cytokine, interacts with CXCR4, a major chemokine receptor, as well as with CD74/invariant chain, a single-pass type II receptor. In this study, we analyzed the roles of CXCR4, CD74, and MIF in CLL. Mononuclear cells from patients with hematological malignancies were analyzed for coexpression of CXCR4 and CD74 by flow cytometry. Strong co- and overexpression of CXCR4 and CD74 were observed on B cells of CLL patients (n = 10). Survival and chemotaxis assays indicated that CXCR4 and CD74 work together to enhance the survival and migration of malignant cells in CLL. Blockade of the receptors, either individually or in combination, promoted cell death and led to an abrogation of MIF-driven migration responses in murine and human CLL cells, suggesting that joint activation of both receptors is crucial for CLL cell survival and mobility. These findings indicate that the MIF/CXCR4/CD74 axis represents a novel therapeutic target in CLL.
Collapse
Affiliation(s)
- Tharshika Thavayogarajah
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, Rheinisch-Westfälische Technische (RWTH) Aachen University, Aachen, Germany; Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany; Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dzmitry Sinitski
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Omar El Bounkari
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Laura Torres-Garcia
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hadas Lewinsky
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Harjung
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hong-Ru Chen
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jens Panse
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, Rheinisch-Westfälische Technische (RWTH) Aachen University, Aachen, Germany
| | - Lucia Vankann
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, Rheinisch-Westfälische Technische (RWTH) Aachen University, Aachen, Germany
| | - Idit Shachar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany; SyNergy Excellence Cluster, Munich, Germany.
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, Rheinisch-Westfälische Technische (RWTH) Aachen University, Aachen, Germany.
| |
Collapse
|
5
|
Klemke L, De Oliveira T, Witt D, Winkler N, Bohnenberger H, Bucala R, Conradi LC, Schulz-Heddergott R. Hsp90-stabilized MIF supports tumor progression via macrophage recruitment and angiogenesis in colorectal cancer. Cell Death Dis 2021; 12:155. [PMID: 33542244 PMCID: PMC7862487 DOI: 10.1038/s41419-021-03426-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/02/2021] [Accepted: 01/07/2021] [Indexed: 12/19/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream regulator of innate immunity, but its expression is increased in some cancers via stabilization with HSP90-associated chaperones. Here, we show that MIF stabilization is tumor-specific in an acute colitis-associated colorectal cancer (CRC) mouse model, leading to tumor-specific functions and selective therapeutic vulnerabilities. Therefore, we demonstrate that a Mif deletion reduced CRC tumor growth. Further, we define a dual role for MIF in CRC tumor progression. Mif deletion protects mice from inflammation-associated tumor initiation, confirming the action of MIF on host inflammatory pathways; however, macrophage recruitment, neoangiogenesis, and proliferative responses are reduced in Mif-deficient tumors once the tumors are established. Thus, during neoplastic transformation, the function of MIF switches from a proinflammatory cytokine to an angiogenesis promoting factor within our experimental model. Mechanistically, Mif-containing tumor cells regulate angiogenic gene expression via a MIF/CD74/MAPK axis in vitro. Clinical correlation studies of CRC patients show the shortest overall survival for patients with high MIF levels in combination with CD74 expression. Pharmacological inhibition of HSP90 to reduce MIF levels decreased tumor growth in vivo, and selectively reduced the growth of organoids derived from murine and human tumors without affecting organoids derived from healthy epithelial cells. Therefore, novel, clinically relevant Hsp90 inhibitors provide therapeutic selectivity by interfering with tumorigenic MIF in tumor epithelial cells but not in normal cells. Furthermore, Mif-depleted colonic tumor organoids showed growth defects compared to wild-type organoids and were less susceptible toward HSP90 inhibitor treatment. Our data support that tumor-specific stabilization of MIF promotes CRC progression and allows MIF to become a potential and selective therapeutic target in CRC.
Collapse
Affiliation(s)
- Luisa Klemke
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Daria Witt
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Winkler
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Richard Bucala
- Departments of Medicine, Pathology, and Epidemiology & Public Health, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | | |
Collapse
|
6
|
Kang I, Bucala R. The immunobiology of MIF: function, genetics and prospects for precision medicine. Nat Rev Rheumatol 2019; 15:427-437. [DOI: 10.1038/s41584-019-0238-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 01/01/2023]
|
7
|
Brocks T, Fedorchenko O, Schliermann N, Stein A, Moll UM, Seegobin S, Dewor M, Hallek M, Marquardt Y, Fietkau K, Heise R, Huth S, Pfister H, Bernhagen J, Bucala R, Baron JM, Fingerle-Rowson G. Macrophage migration inhibitory factor protects from nonmelanoma epidermal tumors by regulating the number of antigen-presenting cells in skin. FASEB J 2016; 31:526-543. [PMID: 27825106 DOI: 10.1096/fj.201600860r] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/04/2016] [Indexed: 12/29/2022]
Abstract
The response of the skin to harmful environmental agents is shaped decisively by the status of the immune system. Keratinocytes constitutively express and secrete the chemokine-like mediator, macrophage migration inhibitory factor (MIF), more strongly than dermal fibroblasts, thereby creating a MIF gradient in skin. By using global and epidermis-restricted Mif-knockout (Mif-/- and K14-Cre+/tg; Miffl/fl) mice, we found that MIF both recruits and maintains antigen-presenting cells in the dermis/epidermis. The reduced presence of antigen-presenting cells in the absence of MIF was associated with accelerated and increased formation of nonmelanoma skin tumors during chemical carcinogenesis. Our results demonstrate that MIF is essential for maintaining innate immunity in skin. Loss of keratinocyte-derived MIF leads to a loss of control of epithelial skin tumor formation in chemical skin carcinogenesis, which highlights an unexpected tumor-suppressive activity of MIF in murine skin.-Brocks, T., Fedorchenko, O., Schliermann, N., Stein, A., Moll, U. M., Seegobin, S., Dewor, M., Hallek, M., Marquardt, Y., Fietkau, K., Heise, R., Huth, S., Pfister, H., Bernhagen, J., Bucala, R., Baron, J. M., Fingerle-Rowson, G. Macrophage migration inhibitory factor protects from nonmelanoma epidermal tumors by regulating the number of antigen-presenting cells in skin.
Collapse
Affiliation(s)
- Tania Brocks
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Oleg Fedorchenko
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Nicola Schliermann
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Astrid Stein
- Institute of Pathology and Cytology, University Hospital Cologne, Cologne, Germany
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, New York, USA.,Department of Molecular Oncology, Georg-August University, Göttingen Center of Molecular Biosciences, Ernst-Caspari-Haus, Göttingen, Germany
| | - Seth Seegobin
- Department of Medical and Molecular Genetics, School of Medicine, Guy's Hospital, King's College London, London, United Kingdom
| | - Manfred Dewor
- Institute of Biochemistry and Molecular Cell Biology Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany.,Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Yvonne Marquardt
- Department of Dermatology, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Katharina Fietkau
- Department of Dermatology, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Ruth Heise
- Department of Dermatology, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Sebastian Huth
- Department of Dermatology, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Herbert Pfister
- Institute of Virology, University Hospital Cologne, Cologne, Germany
| | - Juergen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology Rheinisch-Westfälische Technische Hochschule, Aachen, Germany.,Department of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; and
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jens M Baron
- Department of Dermatology, Rheinisch-Westfälische Technische Hochschule, Aachen, Germany
| | - Guenter Fingerle-Rowson
- Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany; .,Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| |
Collapse
|
8
|
Galletti G, Caligaris-Cappio F, Bertilaccio MTS. B cells and macrophages pursue a common path toward the development and progression of chronic lymphocytic leukemia. Leukemia 2016; 30:2293-2301. [DOI: 10.1038/leu.2016.261] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 12/30/2022]
|
9
|
O'Reilly C, Doroudian M, Mawhinney L, Donnelly SC. Targeting MIF in Cancer: Therapeutic Strategies, Current Developments, and Future Opportunities. Med Res Rev 2016; 36:440-60. [PMID: 26777977 DOI: 10.1002/med.21385] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/28/2015] [Accepted: 10/26/2015] [Indexed: 12/16/2022]
Abstract
Strong evidence has been presented linking chronic inflammation to the onset and pathogenesis of cancer. The multifunctional pro-inflammatory protein macrophage migration inhibitory factor (MIF) occupies a central role in the inflammatory pathway and has been implicated in the tumorigenesis, angiogenesis, and metastasis of many cancer phenotypes. This review highlights the current state of the art, which presents MIF, and the second member of the MIF structural superfamily, D-DT (MIF2), as significant mediators in the inflammatory-cancer axis. Although the mechanism by which MIF asserts its biological activity has yet to be fully understood, it has become clear in recent years that for certain phenotypes of cancer, MIF represents a valid therapeutic target. Current research efforts have focused on small molecule approaches that target MIF's unique tautomerase active site and neutralization of MIF with anti-MIF antibodies. These approaches have yielded promising results in a number of preclinical murine cancer models and have helped to increase our understanding of MIF biological activity. More recently, MIF's involvement in a number of key protein-protein interactions, such as with CD74 and HSP90, has been highlighted and provides a novel platform for the development of anti-MIF chemotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Ciaran O'Reilly
- Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Mohammad Doroudian
- Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Leona Mawhinney
- Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Seamas C Donnelly
- Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.,Department of Clinical Medicine, Trinity Centre for Health Sciences, Tallaght Hospital, Tallaght, Dublin 24, Ireland
| |
Collapse
|
10
|
Yoon K. Gastric Cancer: H. pylori and Macrophage Migration Inhibitory Factor. HELICOBACTER PYLORI 2016:269-274. [DOI: 10.1007/978-981-287-706-2_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Xia W, Xie C, Jiang M, Hou M. Improved survival of mesenchymal stem cells by macrophage migration inhibitory factor. Mol Cell Biochem 2015; 404:11-24. [PMID: 25701358 PMCID: PMC4544672 DOI: 10.1007/s11010-015-2361-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/14/2015] [Indexed: 12/22/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a critical inflammatory cytokine that was recently associated with progenitor cell survival and potently inhibits apoptosis. We examined the protective effect of MIF on hypoxia/serum deprivation (SD)-induced apoptosis of mesenchymal stem cells (MSCs), as well as the possible mechanisms. MSCs were obtained from rat bone marrow and cultured in vitro. Apoptosis was induced by culturing MSCs under hypoxia/SD conditions for up to 24 h and assessed by flow cytometry. Expression levels of c-Met, Akt, and FOXO3a were detected by Western blotting. CD74 expression was detected by qRT-PCR, Western blot, and immunofluorescence. Oxidative stress under hypoxia/SD was examined by detection of reactive oxygen species (ROS) and activity of superoxide dismutase (SOD) and malondialdehyde (MDA). Hypoxia/SD-induced apoptosis was significantly attenuated by recombinant rat MIF in a concentration-dependent manner. MIF induced CD74-asssociated c-Met activation, which was blocked by knocking down CD74 expression using siRNA. MIF also induced Akt and associated FOXO3a phosphorylation, and this effect was abolished by knocking down either CD74 or Akt. In addition, MIF decreased oxidative stress in MSCs, as shown by decreased ROS and MDA, and increased the activity of SOD. Knockdown of CD74, Akt, or FOXO3a largely attenuated the anti-apoptotic effect of MIF and its ability to protect against oxidative stress. MIF protected MSCs from hypoxia/SD-induced apoptosis by interacting with CD74 to stimulate c-Met, leading to downstream PI3K/Akt-FOXO3a signaling and decreased oxidative stress.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | | | | | | |
Collapse
|
12
|
Targeting the heat shock protein 90: a rational way to inhibit macrophage migration inhibitory factor function in cancer. Curr Opin Oncol 2014; 26:108-13. [PMID: 24225413 DOI: 10.1097/cco.0000000000000036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Macrophage migration inhibitory factor (MIF), originally identified as a proinflammatory cytokine, is highly elevated in many human cancer types, independent of their histological origin. MIF's tumour promoting activities correlate with tumour aggressiveness and poor clinical prognosis. Genetic depletion of MIF in mouse cancer models results in significant inhibition of cell proliferation and induction of apoptosis, making it an attractive target for anticancer therapies. Here, we summarize the current possibilities to inhibit MIF function in cancer. RECENT FINDINGS All known small molecule MIF inhibitors antagonize MIF's enzymatic function. However, a recent knockin mouse model suggested that protein interactions play a bigger biological role in tumour cell growth regulation than MIF's enzymatic activity. Thus, alternative strategies are important for targeting MIF. Recently, we identified that MIF in cancer cells is highly stabilized through the heat shock protein 90 machinery (HSP90). Thus, MIF is an HSP90 client. Pharmacological inhibition of the Hsp90 ATPase activity results in MIF degradation in several types of cancer cells. This provides a new way to inhibit MIF function independent of its enzymatic activity. SUMMARY Targeting the HSP90 machinery is a promising way to inhibit MIF function in cancer. Along with MIF and dependent on the molecular make-up of the tumour, a large number of other critical tumourigenic proteins are also destabilized by HSP90 inhibition, overall resulting in a profound block of tumour growth.
Collapse
|
13
|
HER2/ErbB2 activates HSF1 and thereby controls HSP90 clients including MIF in HER2-overexpressing breast cancer. Cell Death Dis 2014; 5:e980. [PMID: 24384723 PMCID: PMC4040658 DOI: 10.1038/cddis.2013.508] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/30/2013] [Accepted: 11/11/2013] [Indexed: 01/31/2023]
Abstract
Overexpression of the human epidermal growth factor receptor-2 (HER2) in breast cancer strongly correlates with aggressive tumors and poor prognosis. Recently, a positive correlation between HER2 and MIF (macrophage migration inhibitory factor, a tumor-promoting protein and heat-shock protein 90 (HSP90) client) protein levels was shown in cancer cells. However, the underlying mechanistic link remained unknown. Here we show that overexpressed HER2 constitutively activates heat-shock factor 1 (HSF1), the master transcriptional regulator of the inducible proteotoxic stress response of heat-shock chaperones, including HSP90, and a crucial factor in initiation and maintenance of the malignant state. Inhibiting HER2 pharmacologically by Lapatinib (a dual HER2/epidermal growth factor receptor inhibitor) or CP724.714 (a specific HER2 inhibitor), or by knockdown via siRNA leads to inhibition of phosphoactivated Ser326 HSF1, and subsequently blocks the activity of the HSP90 chaperone machinery in HER2-overexpressing breast cancer lines. Consequently, HSP90 clients, including MIF, AKT, mutant p53 and HSF1 itself, become destabilized, which in turn inhibits tumor proliferation. Mechanistically, HER2 signals via the phosphoinositide-3-kinase (PI3K)–AKT– mammalian target of rapamycin (mTOR) axis to induce activated pSer326 HSF1. Heat-shock stress experiments confirm this functional link between HER2 and HSF1, as HER2 (and PI3K) inhibition attenuate the HSF1-mediated heat-shock response. Importantly, we confirmed this axis in vivo. In the mouse model of HER2-driven breast cancer, ErbB2 inhibition by Lapatinib strongly suppresses tumor progression, and this is associated with inactivation of the HSF1 pathway. Moreover, ErbB2-overexpressing cancer cells derived from a primary mouse ErbB2 tumor also show HSF1 inactivation and HSP90 client destabilization in response to ErbB2 inhibition. Furthermore, in HER2-positive human breast cancers HER2 levels strongly correlate with pSer326 HSF1 activity. Our results show for the first time that HER2/ErbB2 overexpression controls HSF1 activity, with subsequent stabilization of numerous tumor-promoting HSP90 clients such as MIF, AKT and HSF1 itself, thereby causing a robust promotion in tumor growth in HER2-positive breast cancer.
Collapse
|
14
|
Babu SN, Chetal G, Kumar S. Macrophage migration inhibitory factor: a potential marker for cancer diagnosis and therapy. Asian Pac J Cancer Prev 2013; 13:1737-44. [PMID: 22901113 DOI: 10.7314/apjcp.2012.13.5.1737] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pluripotent cytokine which plays roles in inflammation, immune responses and cancer development. It assists macrophages in carrying out functions like phagocytosis, adherence and motility. Of late, MIF is implicated in almost all stages of neoplasia and expression is a feature of most types of cancer. The presence of MIF in almost all tumors and all stages of cancer makes it an interesting candidate for cancer therapy. This review explores the roles of MIF in neoplasia.
Collapse
Affiliation(s)
- Spoorthy N Babu
- IGNOU-I2IT Centre of Excellence for Advanced Education and Research, Pune, Maharashtra, India
| | | | | |
Collapse
|
15
|
Delayed development of chronic lymphocytic leukemia in the absence of macrophage migration inhibitory factor. Blood 2012; 121:812-21. [PMID: 23118218 DOI: 10.1182/blood-2012-05-431452] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Survival of chronic lymphocytic leukemia (CLL) cells depends on stimuli provided by a suitable microenvironment. The factors and mechanisms providing this growth support for CLL cells are not fully understood. We found that plasma levels of macrophage migration inhibitory factor (MIF), a proinflammatory and immunoregulatory chemokine, were elevated in CLL patients. Therefore, we characterized the functional role of MIF in a CLL mouse model. For this purpose, we crossed Eμ-TCL1 mice with MIF knockout (MIF-/-) mice. The resulting TCL1+/wtMIF/ mice showed a delayed onset of leukemia, reduced splenomegaly and hepatomegaly, and a longer survival than TCL1+/wtMIFwt/wt controls. Immunohistochemical examination of the lymphoid organs showed that the numbers of macrophages were significantly reduced in the spleen and bone marrow of TCL1+/wtMIF/ mice compared with TCL1+/wtMIFwt/wt controls. Mechanistic studies in vitro revealed that the absence of MIF rendered CLL cells more susceptible to apoptosis. Accordingly, incubation with an anti-MIF antibody reduced the survival of CLL cells on a macrophage feeder layer. In addition, the migratory activity of TCL1+/wtMIF/ macrophages was decreased compared with TCL1+/wtMIFwt/wt macrophages. Taken together, our results provide evidence that MIF supports the development of CLL by enhancing the interaction of CLL cells with macrophages. KEY POINTS Targeted deletion of the gene for macrophage migration inhibitory factor (MIF) delays development of chronic lymphocytic leukemia and prolongs survival in mice. MIF recruits leukemia-associated macrophages to spleen or liver.
Collapse
|
16
|
Brain miffed by macrophage migration inhibitory factor. Int J Cell Biol 2012; 2012:139573. [PMID: 22973314 PMCID: PMC3438795 DOI: 10.1155/2012/139573] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/06/2012] [Accepted: 07/12/2012] [Indexed: 12/31/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine which also exhibits enzymatic properties like oxidoreductase and tautomerase. MIF plays a pivotal role in innate and acquired immunity as well as in the neuroendocrine axis. Since it is involved in the pathogenesis of acute and chronic inflammation, neoangiogenesis, and cancer, MIF and its signaling components are considered suitable targets for therapeutic intervention in several fields of medicine. In neurodegenerative and neurooncological diseases, MIF is a highly relevant, but still a hardly investigated mediator. MIF operates via intracellular protein-protein interaction as well as in CD74/CXCR2/CXCR4 receptor-mediated pathways to regulate essential cellular systems such as redox balance, HIF-1, and p53-mediated senescence and apoptosis as well as multiple signaling pathways. Acting as an endogenous glucocorticoid antagonist, MIF thus represents a relevant resistance gene in brain tumor therapies. Alongside this dual action, a functional homolog-annotated D-dopachrome tautomerase/MIF-2 has been uncovered utilizing the same cell surface receptor signaling cascade as MIF. Here we review MIF actions with respect to redox regulation in apoptosis and in tumor growth as well as its extracellular function with a focus on its potential role in brain diseases. We consider the possibility of MIF targeting in neurodegenerative processes and brain tumors by novel MIF-neutralizing approaches.
Collapse
|
17
|
Schulz R, Marchenko ND, Holembowski L, Fingerle-Rowson G, Pesic M, Zender L, Dobbelstein M, Moll UM. Inhibiting the HSP90 chaperone destabilizes macrophage migration inhibitory factor and thereby inhibits breast tumor progression. ACTA ACUST UNITED AC 2012; 209:275-89. [PMID: 22271573 PMCID: PMC3280870 DOI: 10.1084/jem.20111117] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
In several human cancer cell lines, HSP90 inhibitors destabilize macrophage inhibitory factor protein; systemic treatment with an HSP90 inhibitor slows tumor growth and extends overall survival in a mouse model of HER2-positive human breast cancer. Intracellular macrophage migration inhibitory factor (MIF) often becomes stabilized in human cancer cells. MIF can promote tumor cell survival, and elevated MIF protein correlates with tumor aggressiveness and poor prognosis. However, the molecular mechanism facilitating MIF stabilization in tumors is not understood. We show that the tumor-activated HSP90 chaperone complex protects MIF from degradation. Pharmacological inhibition of HSP90 activity, or siRNA-mediated knockdown of HSP90 or HDAC6, destabilizes MIF in a variety of human cancer cells. The HSP90-associated E3 ubiquitin ligase CHIP mediates the ensuing proteasome-dependent MIF degradation. Cancer cells contain constitutive endogenous MIF–HSP90 complexes. siRNA-mediated MIF knockdown inhibits proliferation and triggers apoptosis of cultured human cancer cells, whereas HSP90 inhibitor-induced apoptosis is overridden by ectopic MIF expression. In the ErbB2 transgenic model of human HER2-positive breast cancer, genetic ablation of MIF delays tumor progression and prolongs overall survival of mice. Systemic treatment with the HSP90 inhibitor 17AAG reduces MIF expression and blocks growth of MIF-expressing, but not MIF-deficient, tumors. Together, these findings identify MIF as a novel HSP90 client and suggest that HSP90 inhibitors inhibit ErbB2-driven breast tumor growth at least in part by destabilizing MIF.
Collapse
Affiliation(s)
- Ramona Schulz
- Department of Molecular Oncology, Göttingen Center of Molecular Biosciences, University of Göttingen, 37077 Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Xiao DZ, Dai B, Chen J, Luo Q, Liu XY, Lin QX, Li XH, Huang W, Yu XY. Loss of macrophage migration inhibitory factor impairs the growth properties of human HeLa cervical cancer cells. Cell Prolif 2011; 44:582-90. [PMID: 21991924 DOI: 10.1111/j.1365-2184.2011.00787.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES This study aims to determine the role of macrophage migration inhibitory factor (MIF), a proinflammatory cytokine associated with cell proliferation and tumour growth in vivo. MATERIALS AND METHODS Our team used RNA interference technology to knock down MIF expression in human HeLa cervical cancer cells and to establish a stable cell line lacking MIF function. RESULTS Our results showed that long-term loss of MIF had little effect on cell morphology, but significantly inhibited their population growth and proliferation. The HeLa MIF-knockdown cells retained normal apoptotic signalling pathways in response to TNF-alpha treatment; however, they exhibited unique DNA profiles following doxorubicin treatment, suggesting that MIF may regulate a cell cycle checkpoint upon DNA damage. Our data also showed that knockdown of MIF expression in HeLa cells led to increased cell adhesion and therefore impaired their migratory capacity. More importantly, cells lacking MIF failed to either proliferate in soft agar or form tumours in vivo, when administered to nude mice. CONCLUSION MIF plays a pivotal role in proliferation and tumourigenesis of human HeLa cervical carcinoma cells, and may represent a promising therapeutic target for cancer intervention.
Collapse
Affiliation(s)
- D Z Xiao
- Medical Research Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
G to C transition at position -173 of MIF gene of the recipient is associated with reduced relapse rates after allogeneic stem cell transplantation. Cytokine 2009; 48:218-25. [PMID: 19720544 DOI: 10.1016/j.cyto.2009.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2009] [Revised: 06/30/2009] [Accepted: 07/20/2009] [Indexed: 11/22/2022]
Abstract
Pro-inflammatory and dendritic cell-activating properties of macrophage migration inhibitory factor (MIF) suggest a potentially important role for MIF in alloantigen-specific immune responses after allogeneic stem cell transplantation (allo-SCT). We tested whether MIF -173 G/C gene polymorphism of donor or patient had impacts on the outcomes after allo-SCT. Four hundred and fifty-four donor-patient pairs were genotyped and mortality, relapse, and development of complications were analyzed. Patient but not donor MIF -173 *C allele was associated with improved overall survival (OS) (5 years: 60.8% versus 46.3%, p=0.042) and disease free survival (DFS) (5 years: 55.4% versus 39.5%; p=0.014) due to a reduction in relapse (day 2000: 22.8% versus 42.0% p=0.006) but not due to decreased transplantation-related mortality (TRM) (p=0.44). Multivariate analysis proved patient -173 *C allele as an independent factor for reducing relapse after allo-SCT (p=0.023). Subgroup analysis showed a clear MIF -173 *C allele-related reduction in relapse for those patients who did not receive T cell depleted (TCD) SCT (p=0.01) in contrast to patients receiving TCD SCT (p=0.20). In summary, patient MIF -173 *C allele may be linked to specific, yet unrevealed functions in tumor biology and graft versus leukemia and lymphoma effects and potentially presents a novel prognostic marker for patient-tailored counseling and therapy in allo-SCT.
Collapse
|
20
|
Li YY, Hsieh LL, Tang RP, Liao SK, Yeh KY. Macrophage-derived interleukin-6 up-regulates MUC1, but down-regulates MUC2 expression in the human colon cancer HT-29 cell line. Cell Immunol 2009; 256:19-26. [PMID: 19201396 DOI: 10.1016/j.cellimm.2009.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 12/22/2008] [Accepted: 01/02/2009] [Indexed: 02/07/2023]
Abstract
Little is known regarding the effects of IL-6 released by tumor-infiltrating macrophages on the mucin expression of colon cancer cells. We isolated macrophages from healthy donors and harvested the supernatant after 48-h cultures. Using flow cytometry and intracellular staining methods, we found that macrophage supernatant effectively induced MUC1 up-regulation and MUC2 down-regulation of colon cancer cells in vitro. Western blotting analysis using monoclonal antibody against IL-6 or gp 130 verified that this IL-6-driven activity was through the activation of tyrosine phosphorylation (Tyr(705)) of STAT3 in cancer cells. We analyzed the surgical specimens of 29 patients with colon cancer by an immunohistochemical staining method and demonstrated the co-localization of macrophages, and the expression of IL-6, CD68, and MUC1 in colon cancer patients. Therefore, macrophage-derived IL-6 modulates the mucin expression of colorectal cancer cells that might in turn produce a permissive milieu favorable to cancer spread.
Collapse
Affiliation(s)
- Ying-Ying Li
- Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung & Chang Gung University, Taiwan
| | | | | | | | | |
Collapse
|
21
|
A tautomerase-null macrophage migration-inhibitory factor (MIF) gene knock-in mouse model reveals that protein interactions and not enzymatic activity mediate MIF-dependent growth regulation. Mol Cell Biol 2009; 29:1922-32. [PMID: 19188446 DOI: 10.1128/mcb.01907-08] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Macrophage migration-inhibitory factor (MIF) is an upstream regulator of innate immunity and a potential molecular link between inflammation and cancer. The unusual structural homology between MIF and certain tautomerases, which includes both a conserved substrate-binding pocket and a catalytic N-terminal proline (Pro1), has fueled speculation that an enzymatic reaction underlies MIF's biologic function. To address the functional role of the MIF tautomerase activity in vivo, we created a knock-in mouse in which the endogenous mif gene was replaced by one encoding a tautomerase-null, Pro1-->Gly1 MIF protein (P1G-MIF). While P1G-MIF is completely inactive catalytically, it maintains significant, albeit reduced, binding to its cell surface receptor (CD74) and to the intracellular binding protein JAB1/CSN5. P1G-MIF knock-in mice (mif(P1G/P1G)) and cells derived from these mice show a phenotype in assays of growth control and tumor induction that is intermediate between those of the wild type (mif(+/+)) and complete MIF deficiency (mif(-)(/)(-)). These data provide genetic evidence that MIF's intrinsic tautomerase activity is dispensable for this cytokine's growth-regulatory properties and support a role for the N-terminal region in protein-protein interactions.
Collapse
|
22
|
Stein R, Mattes MJ, Cardillo TM, Hansen HJ, Chang CH, Burton J, Govindan S, Goldenberg DM. CD74: a new candidate target for the immunotherapy of B-cell neoplasms. Clin Cancer Res 2007; 13:5556s-5563s. [PMID: 17875789 DOI: 10.1158/1078-0432.ccr-07-1167] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CD74 is an integral membrane protein that functions as a MHC class II chaperone. Moreover, it has recently been shown to have a role as an accessory-signaling molecule and has been implicated in malignant B-cell proliferation and survival. These biological functions combined with expression of CD74 on malignant B cells and limited expression on normal tissues implicate CD74 as a potential therapeutic target. The anti-CD74 monoclonal antibody LL1 has been humanized (hLL1 milatuzumab or IMMU-115) and can provide the basis for novel therapeutic approaches to B-cell malignancies, particularly because this antibody shows rapid internalization into CD74+ malignant cells. This article reviews the preclinical evaluations of LL1, its humanized form, and isotope, drug, and toxin conjugates. These studies show that unconjugated hLL1 and conjugates of hLL1 constructs with radioisotopes, doxorubicin, and frog RNase have high antitumor activity in non-Hodgkin's lymphoma and multiple myeloma in vitro and in tumor xenograft models. Single-dose studies of hLL1 in monkeys showed no adverse effects but did decrease circulating B and T lymphocytes and natural killer cells. When evaluated in combination with rituximab, either equivalent or improved efficacy, compared with either antibody alone, was observed. CD74 is a new candidate target for the immunotherapy of neoplasms expressing this antigen, which can be exploited using either a naked antibody or conjugated to isotopes, drugs, or toxins.
Collapse
Affiliation(s)
- Rhona Stein
- Garden State Cancer Center, Center for Molecular Medicine and Immunology, Belleville, New Jersey 07109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Willeke P, Gaubitz M, Schotte H, Maaser C, Domschke W, Schlüter B, Becker H. Increased serum levels of macrophage migration inhibitory factor in patients with primary Sjögren's syndrome. Arthritis Res Ther 2007; 9:R43. [PMID: 17470266 PMCID: PMC1906791 DOI: 10.1186/ar2182] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/16/2007] [Accepted: 04/30/2007] [Indexed: 11/21/2022] Open
Abstract
The objective of this study was to analyse levels of the proinflammatory cytokine macrophage migration inhibitory factor (MIF) in patients with primary Sjögren's syndrome (pSS) and to examine associations of MIF with clinical, serological and immunological variables. MIF was determined by ELISA in the sera of 76 patients with pSS. Further relevant cytokines (IL-1, IL-6, IL-10, IFN-γ and TNF-α) secreted by peripheral blood mononuclear cells (PBMC) were determined by ELISPOT assay. Lymphocytes and monocytes were examined flow-cytometrically for the expression of activation markers. Results were correlated with clinical and laboratory findings as well as with the HLA-DR genotype. Healthy age- and sex-matched volunteers served as controls. We found that MIF was increased in patients with pSS compared with healthy controls (p < 0.01). In particular, increased levels of MIF were associated with hypergammaglobulinemia. Further, we found a negative correlation of MIF levels with the number of IL-10-secreting PBMC in pSS patients (r = -0.389, p < 0.01). Our data indicate that MIF might participate in the pathogenesis of primary Sjögren's syndrome. MIF may contribute to B-cell hyperactivity indicated by hypergammaglobulinemia. The inverse relationship of IL-10 and MIF suggests that IL-10 works as an antagonist of MIF in pSS.
Collapse
Affiliation(s)
- Peter Willeke
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Markus Gaubitz
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Heiko Schotte
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Christian Maaser
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Wolfram Domschke
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Bernhard Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Heidemarie Becker
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| |
Collapse
|
24
|
Fingerle-Rowson G, Petrenko O. MIF coordinates the cell cycle with DNA damage checkpoints. Lessons from knockout mouse models. Cell Div 2007; 2:22. [PMID: 17640378 PMCID: PMC1941730 DOI: 10.1186/1747-1028-2-22] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 07/19/2007] [Indexed: 12/22/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a ubiquitously expressed pro-inflammatory mediator that has also been implicated in the process of oncogenic transformation and tumor progression. We used a genetic approach to show that deletion of the MIF gene in mice has several major consequences for the proliferative and transforming properties of cells. MIF-deficient cells exhibit increased resistance to oncogenic transformation. The transformation defects associated with MIF deficiency can be overcome through concomitant inactivation of the p53 and Rb/E2F tumor suppressor pathways. We have produced compelling evidence that the effects of MIF on cell survival and tumorigenesis are mediated through overlapping pathways, wherein MIF and p53 functionally antagonize each other in the cell. However, the involvement of MIF in p53 function is secondary to p53-independent mechanisms controlling protein stability, DNA damage checkpoints, and the integrity of the genome. Given the broad spectrum of cell types that normally express MIF and its elevated levels at sites of chronic inflammation, this pathway may be generic for many early stage tumors.
Collapse
Affiliation(s)
- Günter Fingerle-Rowson
- University Hospital Cologne, Clinic I of Internal Medicine, Dept. of Hematology and Oncology, LFI, Level 4, Room 704, Kerpenerstr. 62, 50924 Cologne, Germany
| | - Oleksi Petrenko
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| |
Collapse
|
25
|
Bucala R, Donnelly SC. Macrophage migration inhibitory factor: a probable link between inflammation and cancer. Immunity 2007; 26:281-5. [PMID: 17376392 DOI: 10.1016/j.immuni.2007.03.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The pleiotropic effects of macrophage migration inhibitory factor (MIF) place it in a central position in the immunopathogenesis of many diseases. Here we discuss the current understanding of MIF's role and highlight it as a potential link between inflammatory activation and malignant progression.
Collapse
Affiliation(s)
- Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|
26
|
Nemajerova A, Mena P, Fingerle-Rowson G, Moll UM, Petrenko O. Impaired DNA damage checkpoint response in MIF-deficient mice. EMBO J 2007; 26:987-97. [PMID: 17290223 PMCID: PMC1852846 DOI: 10.1038/sj.emboj.7601564] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Accepted: 12/20/2006] [Indexed: 11/09/2022] Open
Abstract
Recent studies demonstrated that proinflammatory migration inhibitory factor(MIF) blocks p53-dependent apoptosis and interferes with the tumor suppressor activity of p53. To explore the mechanism underlying this MIF-p53 relationship, we studied spontaneous tumorigenesis in genetically matched p53-/- and MIF-/-p53-/- mice. We show that the loss of MIF expression aggravates the tumor-prone phenotype of p53-/- mice and predisposes them to a broader tumor spectrum, including B-cell lymphomas and carcinomas. Impaired DNA damage response is at the root of tumor predisposition of MIF-/-p53-/- mice. We provide evidence that MIF plays a role in regulating the activity of Cul1-containing SCF ubiquitin ligases. The loss of MIF expression uncouples Chk1/Chk2-responsive DNA damage checkpoints from SCF-dependent degradation of key cell-cycle regulators such as Cdc25A, E2F1 and DP1, creating conditions for the genetic instability of cells. These MIF effects depend on its association with the Jab1/CSN5 subunit of the COP9/CSN signalosome. Given that CSN plays a central role in the assembly of SCF complexes in vivo, regulation of Jab1/CSN5 by MIF is required to sustain optimal composition and function of the SCF complex.
Collapse
Affiliation(s)
- Alice Nemajerova
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Patricio Mena
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Gunter Fingerle-Rowson
- University Hospital Cologne, Medical Clinic I, Hematology and Oncology, Cologne, Germany
| | - Ute M Moll
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Oleksi Petrenko
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, NY, USA
- Department of Pathology, State University of New York at Stony Brook, BST L9, Stony Brook, NY 11794, USA. Tel.: +1 631 444 3520; Fax: +1 631 444 3424; E-mail:
| |
Collapse
|
27
|
Welford SM, Bedogni B, Gradin K, Poellinger L, Broome Powell M, Giaccia AJ. HIF1alpha delays premature senescence through the activation of MIF. Genes Dev 2006; 20:3366-71. [PMID: 17142669 PMCID: PMC1698444 DOI: 10.1101/gad.1471106] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Premature senescence in vitro has been attributed to oxidative stress leading to a DNA damage response. In the absence of oxidative damage that occurs at atmospheric oxygen levels, proliferation of untransformed cells continues for extended periods of time. We have investigated the role of the hypoxia-inducible factor 1alpha (HIF1alpha) transcription factor in preventing senescence in aerobic and hypoxic conditions. Using embryonic fibroblasts from a conditional HIF1alpha knockout mouse, we found that loss of HIF1alpha under aerobic conditions significantly accelerated the onset of cellular senescence, and decreased proliferation under hypoxia. Furthermore, we identify the macrophage migration inhibitory factor (MIF) as a crucial effector of HIF1alpha that delays senescence. Inhibition of MIF phenocopies loss of HIF1alpha. Our findings highlight a novel role for HIF1alpha under aerobic conditions, and identify MIF as a target responsible for this function.
Collapse
Affiliation(s)
- Scott M. Welford
- Division of Radiation and Cancer Biology, Stanford University, Stanford, California 94305, USA
| | - Barbara Bedogni
- Division of Radiation and Cancer Biology, Stanford University, Stanford, California 94305, USA
| | - Katarina Gradin
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Lorenz Poellinger
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Marianne Broome Powell
- Division of Radiation and Cancer Biology, Stanford University, Stanford, California 94305, USA
| | - Amato J. Giaccia
- Division of Radiation and Cancer Biology, Stanford University, Stanford, California 94305, USA
- Corresponding author.E-MAIL ; FAX (650) 723-7382
| |
Collapse
|
28
|
Abstract
The discovery that the Myc oncoprotein could drive cells to undergo apoptosis in addition to its well-established role in cellular proliferation came in the early 1990s, at the beginning of a period of explosive research on cell death. Experimental evidence revealed that Myc sensitises cells to a wide range of death stimuli and abrogating this biological activity plays a profound role in tumorigenesis. Our understanding of the molecular mechanism and genetic programme of Myc-induced apoptosis remains shrouded in mystery and the focus of much attention. In this review, we will discuss established data, recent advances and future objectives regarding the regulatory processes and the functional cooperators that effect and abrogate apoptosis induced by Myc.
Collapse
Affiliation(s)
- Natalie Meyer
- Division of Cancer Genomics and Proteomics, Ontario Cancer Institute/Princess Margaret Hospital, Department of Medical Biophysics, University of Toronto, Toronto, Ont, Canada
| | | | | |
Collapse
|