1
|
Urbanová V, Lu S, Kalinová E, Martins L, Kozelková T, Dyčka F, Ribeiro JM, Hajdušek O, Perner J, Kopáček P. From the fat body to the hemolymph: Profiling tick immune and storage proteins through transcriptomics and proteomics. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 165:104072. [PMID: 38185274 DOI: 10.1016/j.ibmb.2024.104072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/09/2024]
Abstract
Ticks are blood-feeding arachnids that are known to transmit various pathogenic microorganisms to their hosts. During blood feeding, ticks activate their metabolism and immune system to efficiently utilise nutrients from the host's blood and complete the feeding process. In contrast to insects, in which the fat body is known to be a central organ that controls essential metabolic processes and immune defense mechanisms, the function of the fat body in tick physiology is still relatively unexplored. To fill this gap, we sought to uncover the repertoire of genes expressed in the fat body associated with trachea (FB/Tr) by analyzing the transcriptome of individual, partially fed (previtellogenic) Ixodes ricinus females. The resulting catalog of individual mRNA sequences reveals a broad repertoire of transcripts encoding proteins involved in nutrient storage and distribution, as well as components of the tick immune system. To gain a detailed insight into the secretory products of FB/Tr specifically involved in inter-tissue transport and humoral immunity, the transcriptomic data were complemented with the proteome of soluble proteins in the hemolymph of partially fed female ticks. Among these proteins, the hemolipoglyco-carrier proteins were predominant. When comparing immune peptides and proteins from the fat body with those produced by hemocytes, we found that the fat body serves as a unique producer of certain immune components. Finally, time-resolved transcriptional regulation of selected immune transcripts from the FB/Tr was examined in response to experimental challenges with model microbes and analyzed by RT-qPCR. Overall, our data show that the fat body of ticks, similar to insects, is an important metabolic tissue that also plays a remarkable role in immune defense against invading microbes. These findings improve our understanding of tick biology and its impact on the transmission of tick-borne pathogens.
Collapse
Affiliation(s)
- Veronika Urbanová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Stephen Lu
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Eliška Kalinová
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Larissa Martins
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories - NIH/NIAID, Hamilton, MT, USA
| | - Tereza Kozelková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Filip Dyčka
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - José M Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ondřej Hajdušek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Jan Perner
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic.
| |
Collapse
|
2
|
Ladakis DC, Reyes-Mantilla MI, Gadani SP, Mace JW, Dominguez-Penuela SC, Appiah MJ, Smith MD, Bhargava P, Fox RJ, Saidha S, Calabresi PA. Serum macrophage migration inhibitory factor levels predict brain atrophy in people with primary progressive multiple sclerosis. Mult Scler 2024; 30:35-43. [PMID: 37982154 DOI: 10.1177/13524585231213164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a cytokine linked to multiple sclerosis (MS) progression that is thought to be inhibited by ibudilast. SPRINT-MS was a phase 2 placebo-controlled trial of ibudilast in progressive multiple sclerosis (PMS). OBJECTIVE To determine whether baseline MIF levels predict imaging outcomes and assess the effects of ibudilast on serum and cerebrospinal fluid (CSF) MIF levels in people with PMS treated with ibudilast. METHODS Participants in the SPRINT-MS trial were treated with either ibudilast or placebo and underwent brain magnetic resonance imaging (MRI) every 24 weeks over a duration of 96 weeks. MIF was measured in serum and CSF. RESULTS MIF levels were compared with imaging outcomes in 223 participants from the SPRINT-MS study. In the primary progressive multiple sclerosis (PPMS) cohort, males had higher serum (p < 0.001) and CSF (p = 0.01) MIF levels, as compared with females. Higher baseline serum MIF levels in PPMS were associated with faster brain atrophy (beta = -0.113%, 95% confidence interval (CI): -0.204% to -0.021%; p = 0.016). These findings were not observed in secondary progressive multiple sclerosis (SPMS). Ibudilast did not affect either serum or CSF MIF levels. CONCLUSIONS Serum MIF levels were associated with male sex and predicted brain atrophy in PPMS, but not SPMS. Ibudilast did not demonstrate an effect on MIF levels, as compared with placebo, although we cannot exclude a functional effect.
Collapse
Affiliation(s)
- Dimitrios C Ladakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria I Reyes-Mantilla
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sachin P Gadani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jackson W Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mayaa J Appiah
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Dumas F, Mauro M, Vazzana M, Arizza V, Vizzini A. Ciona robusta macrophage migration inhibitory factor (Mif1 and Mif2) genes are differentially regulated in the lipopolysaccharide-challenged pharynx. JOURNAL OF FISH BIOLOGY 2023; 103:727-730. [PMID: 37148434 DOI: 10.1111/jfb.15427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/04/2023] [Indexed: 05/08/2023]
Abstract
The effects of lipopolysaccharide (LPS) on Mif (macrophage migration inhibitory factor) gene expression in the pharynx (haemapoetic tissue) of Ciona robusta were investigated using quantitative reverse-transcription PCR (qRT-PCR) and in situ hybridisation (ISH). To verify the induction of an inflammatory response in the pharynx, a qRT-PCR analysis was performed to evaluate the change in the expression of proinflammatory marker genes such as Mbl, Ptx-like, Tnf-α and Nf-kb, which were shown to be upregulated 1 h post LPS challenge. The change in the expression of the two Mif paralogs in the pharynx was assessed before and after stimulation, and qRT-PCR and ISH results showed that, although Mif2 and Mif2 were expressed in clusters of haemocytes in pharynx vessels, only Mif1 expression increased after LPS stimulation. This indicates that the Mif genes are differently regulated and respond to different ambient inputs that need further analysis.
Collapse
Affiliation(s)
- Francesca Dumas
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Palermo, Italy
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Palermo, Italy
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Palermo, Italy
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Palermo, Italy
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Palermo, Italy
| |
Collapse
|
4
|
Chen J, Guo W, Du P, Cui T, Yang Y, Wang Y, Kang P, Zhang Z, Wang Q, Ye Z, Liu L, Jian Z, Gao T, Bian H, Li S, Li C. MIF inhibition alleviates vitiligo progression by suppressing CD8 + T cell activation and proliferation. J Pathol 2023; 260:84-96. [PMID: 36852981 DOI: 10.1002/path.6073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/18/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023]
Abstract
In vitiligo, autoreactive CD8+ T cells have been established as the main culprit considering its pathogenic role in mediating epidermal melanocyte-specific destruction. Macrophage migration inhibitory factor (MIF) is a pleiotropic molecule that plays a central role in various immune processes including the activation and proliferation of T cells; but whether MIF is intertwined in vitiligo development and progression and its involvement in aberrantly activated CD8+ T cells remains ill-defined. In this study, we found that MIF was overabundant in vitiligo patients and a mouse model for human vitiligo. Additionally, inhibiting MIF ameliorated the disease progression in vitiligo mice, which manifested as less infiltration of CD8+ T cells and more retention of epidermal melanocytes in the tail skin. More importantly, in vitro experiments indicated that MIF-inhibition suppressed the activation and proliferation of CD8+ T cells from the lymph nodes of vitiligo mice, and the effect extended to CD8+ T cells in peripheral blood mononuclear cells of vitiligo patients. Finally, CD8+ T cells derived from MIF-inhibited vitiligo mice also exhibited an impaired capacity for activation and proliferation. Taken together, our results show that MIF might be clinically targetable in vitiligo treatment, and its inhibition might ameliorate vitiligo progression by suppressing autoreactive CD8+ T cell activation and proliferation. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Pengran Du
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Tingting Cui
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yuqi Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yinghan Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Pan Kang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhe Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Qi Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhubiao Ye
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Ling Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, PR China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| |
Collapse
|
5
|
La Paglia L, Vazzana M, Mauro M, Dumas F, Fiannaca A, Urso A, Arizza V, Vizzini A. Transcriptomic and Bioinformatic Analyses Identifying a Central Mif-Cop9-Nf-kB Signaling Network in Innate Immunity Response of Ciona robusta. Int J Mol Sci 2023; 24:ijms24044112. [PMID: 36835523 PMCID: PMC9960688 DOI: 10.3390/ijms24044112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The Ascidian C. robusta is a powerful model for studying innate immunity. LPS induction activates inflammatory-like reactions in the pharynx and the expression of several innate immune genes in granulocyte hemocytes such as cytokines, for instance, macrophage migration inhibitory factors (CrMifs). This leads to intracellular signaling involving the Nf-kB signaling cascade that triggers downstream pro-inflammatory gene expression. In mammals, the COP9 (Constitutive photomorphogenesis 9) signalosome (CSN) complex also results in the activation of the NF-kB pathway. It is a highly conserved complex in vertebrates, mainly engaged in proteasome degradation which is essential for maintaining processes such as cell cycle, DNA repair, and differentiation. In the present study, we used bioinformatics and in-silico analyses combined with an in-vivo LPS exposure strategy, next-generation sequencing (NGS), and qRT-PCR to elucidate molecules and the temporal dynamics of Mif cytokines, Csn signaling components, and the Nf-κB signaling pathway in C. robusta. A qRT-PCR analysis of immune genes selected from transcriptome data revealed a biphasic activation of the inflammatory response. A phylogenetic and STRING analysis indicated an evolutionarily conserved functional link between the Mif-Csn-Nf-kB axis in ascidian C. robusta during LPS-mediated inflammation response, finely regulated by non-coding molecules such as microRNAs (miRNAs).
Collapse
Affiliation(s)
- Laura La Paglia
- Istituto di Calcolo e Reti ad Alte Prestazioni-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Via Archirafi 18, 90128 Palermo, Italy
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Via Archirafi 18, 90128 Palermo, Italy
| | - Francesca Dumas
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Via Archirafi 18, 90128 Palermo, Italy
| | - Antonino Fiannaca
- Istituto di Calcolo e Reti ad Alte Prestazioni-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Alfonso Urso
- Istituto di Calcolo e Reti ad Alte Prestazioni-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Via Archirafi 18, 90128 Palermo, Italy
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche-Università di Palermo, Via Archirafi 18, 90128 Palermo, Italy
- Correspondence:
| |
Collapse
|
6
|
Varyani F, Löser S, Filbey KJ, Harcus Y, Drurey C, Poveda MC, Rasid O, White MPJ, Smyth DJ, Gerbe F, Jay P, Maizels RM. The IL-25-dependent tuft cell circuit driven by intestinal helminths requires macrophage migration inhibitory factor (MIF). Mucosal Immunol 2022; 15:1243-1256. [PMID: 35288645 PMCID: PMC9705247 DOI: 10.1038/s41385-022-00496-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a key innate immune mediator with chemokine- and cytokine-like properties in the inflammatory pathway. While its actions on macrophages are well-studied, its effects on other cell types are less understood. Here we report that MIF is required for expansion of intestinal tuft cells during infection with the helminth Nippostrongylus brasiliensis. MIF-deficient mice show defective innate responses following infection, lacking intestinal epithelial tuft cell hyperplasia or upregulation of goblet cell RELMβ, and fail to expand eosinophil, type 2 innate lymphoid cell (ILC2) and macrophage (M2) populations. Similar effects were observed in MIF-sufficient wild-type mice given the MIF inhibitor 4-IPP. MIF had no direct effect on epithelial cells in organoid cultures, and MIF-deficient intestinal stem cells could generate tuft cells in vitro in the presence of type 2 cytokines. In vivo the lack of MIF could be fully compensated by administration of IL-25, restoring tuft cell differentiation and goblet cell expression of RELM-β, demonstrating its requirement upstream of the ILC2-tuft cell circuit. Both ILC2s and macrophages expressed the MIF receptor CXCR4, indicating that MIF may act as an essential co-factor on both cell types to activate responses to IL-25 in helminth infection.
Collapse
Affiliation(s)
- Fumi Varyani
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Stephan Löser
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kara J Filbey
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Lydia Becker Institute for Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Yvonne Harcus
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Orhan Rasid
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Madeleine P J White
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - François Gerbe
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Philippe Jay
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
7
|
Zhang Y, Lu S, Fan S, Xu L, Jiang X, Wang K, Cai B. Macrophage migration inhibitory factor activates the inflammatory response in joint capsule fibroblasts following post-traumatic joint contracture. Aging (Albany NY) 2021; 13:5804-5823. [PMID: 33601337 PMCID: PMC7950233 DOI: 10.18632/aging.202505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Objectives: Joint capsule fibrosis caused by excessive inflammation leading to post-traumatic joint contracture (PTJC). Fibroblasts trigger inflammation under the challenge of various proinflammatory cytokines. Macrophage migration inhibitory factor (MIF) is a prominent proinflammatory cytokine involved in inflammation- and fibrosis-associated pathophysiology, we investigated the role of MIF in PTJC. Methods: Using rat PTJC model and fibroblast inflammation model, we detected MIF expression in posterior joint capsule. Primary joint capsule fibroblasts (JFs) were used to investigate the effects of MIF on cell proliferation, migration and proinflammatory cytokines production. The mechanism of JF-mediated events was evaluated by qRT-PCR, western blot and immunoprecipitation. We screened the mRNA expression profile to identify gene candidates that mediate the effect of MIF on JFs. Results: MIF increased in posterior joint capsule following PTJC and co-localized with fibroblasts. Injection of MIF inhibitor significantly suppressed joint capsule inflammation and fibrosis. In vitro, MIF promoted JF proliferation, migration, and inflammation by regulating mitogen-activated protein kinase/nuclear factor-κB pathway through coupling with CD74. Transcriptome analysis revealed that lipid metabolism-related factors Pla2g2a, Angptl4, and Sgpp2, downstream of MIF/CD74, were potentially implicated in JF inflammation. Conclusion: MIF/CD74 axis elicited JF inflammation and may provide new therapeutic targets for joint capsule fibrosis in PTJC.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shenji Lu
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuai Fan
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lili Xu
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xin Jiang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kexin Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Bin Cai
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
8
|
Omara II, Pender CM, White MB, Dalloul RA. The Modulating Effect of Dietary Beta-Glucan Supplementation on Expression of Immune Response Genes of Broilers during a Coccidiosis Challenge. Animals (Basel) 2021; 11:ani11010159. [PMID: 33445562 PMCID: PMC7827683 DOI: 10.3390/ani11010159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Avian coccidiosis is the leading parasitic disease in the poultry industry and means to control its damages continue to be explored. This study evaluated the feeding effects of a yeast-derived β-glucan on expression of immune response genes in the spleen, thymus, and bursa of commercial broiler chickens during an Eimeria challenge. The study consisted of two dietary treatments (0% or 0.1% β-glucan) each with or without a coccidiosis challenge. There were significant effects from dietary β-glucan, Eimeria challenge, and their interaction for several gene targets in the spleen, thymus, and bursa on days 10 and 14 of age. Based on the current results, supplementation of dietary β-glucan in Eimeria-challenged birds enhanced and modulated the expression of immune response genes during coccidiosis. Abstract This study investigated the effects of a yeast-derived β-glucan (Auxoferm YGT) supplementation on mRNA expression of immune response genes in the spleen, thymus, and bursa of broiler chickens during a mixed Eimeria infection. Day (d)-old chicks (n = 1440) were fed diets containing 0% or 0.1% YGT. On d 8 post-hatch, half the replicate pens (n = 8) were challenged with a mixed inoculum of E. acervulina, E. maxima, and E. tenella. On d 10 and d 14 post-hatch, the spleen, thymus, and bursa were collected to evaluate mRNA abundance by quantitative real-time PCR. Data were analyzed using PROC GLIMMIX model (2-way interaction) and differences were established by LS-MEANS with significance reported at p ≤ 0.05. In spleen tissues at d 10, expression of interleukin (IL)-10 and inducible nitric oxide synthase (iNOS) were elevated in both 0.1% YGT-fed challenged and non-challenged birds. In thymus tissues at d 14, expression of IL-10, IL-17F, interferon (IFN)-γ, iNOS, and macrophage migration inhibitory factor (MIF) were elevated in challenged birds fed 0.1% YGT. In bursal tissues at d 10 and d 14, expression of IL-10, IFN-γ, iNOS (d 10 only), and MIF were elevated in 0.1% YGT-fed challenged and non-challenged birds. Dietary β-glucan supplementation to chicken diets modulated their immune response to the Eimeria challenge.
Collapse
Affiliation(s)
- Islam I. Omara
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (I.I.O.); (C.M.P.); (M.B.W.)
- Animal and Poultry Division, Department of Animal Production, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Chasity M. Pender
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (I.I.O.); (C.M.P.); (M.B.W.)
| | - Mallory B. White
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (I.I.O.); (C.M.P.); (M.B.W.)
| | - Rami A. Dalloul
- Avian Immunobiology Laboratory, Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, USA; (I.I.O.); (C.M.P.); (M.B.W.)
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
- Correspondence:
| |
Collapse
|
9
|
Arizza V, Bonura A, La Paglia L, Urso A, Pinsino A, Vizzini A. 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. [PMID: 32647255 PMCID: PMC7347617 DOI: 10.1038/s41598-020-68339-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/16/2020] [Indexed: 01/07/2023] Open
Abstract
The close phylogenetic relationship between Ciona robusta and vertebrates makes it a powerful model for studying innate immunity and the evolution of immune genes. To elucidate the nature and dynamics of the immune response, the molecular mechanisms by which bacterial infection is detected and translated into inflammation and how potential pattern recognition receptors (PRRs) are involved in pathogen recognition in tunicate C. robusta (formerly known as Ciona intestinalis), we applied an approach combining bacterial infections, next-generation sequencing, qRT-PCR, bioinformatics and in silico analyses (criteria of a p-value < 0.05 and FDR < 0.05). A STRING analysis indicated a functional link between components of the Tlr/MyD88-dependent signalling pathway (Tlr2, MyD88, and Irak4) and components of the Nf-κB signalling pathway (Nf-κB, IκBα, and Ikkα) (p-value < 0.05, FDR < 0.05). A qRT-PCR analysis of immune genes selected from transcriptome data revealed Mif as more frequently expressed in the inflammatory response than inflammation mediator or effector molecules (e.g., Il-17s, Tnf-α, Tgf-β, Mmp9, Tlrs, MyD88, Irak4, Nf-κB, and galectins), suggesting close interplay between Mif cytokines and Nf-κB signalling pathway components in the biphasic activation of the inflammatory response. An in silico analyses of the 3′-UTR of Tlr2, MyD88, IκBα, Ikk, and Nf-κB transcripts showed the presence of GAIT elements, which are known to play key roles in the regulation of immune gene-specific translation in humans. These findings provide a new level of understanding of the mechanisms involved in the regulation of the C. robusta inflammatory response induced by LPS and suggest that in C. robusta, as in humans, a complex transcriptional and post-transcriptional control mechanism is involved in the regulation of several inflammatory genes.
Collapse
Affiliation(s)
- Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, Via Archirafi 18, Palermo, Italy
| | - Angela Bonura
- Istituto per la Ricerca e l'Innovazione Biomedica-Consiglio Nazionale delle Ricerche, Via Ugo la Malfa 153, Palermo, Italy
| | - Laura La Paglia
- Istituto di Calcolo e Reti ad Alte Prestazioni-Consiglio Nazionale delle Ricerche, Via Ugo la Malfa 153, Palermo, Italy
| | - Alfonso Urso
- Istituto di Calcolo e Reti ad Alte Prestazioni-Consiglio Nazionale delle Ricerche, Via Ugo la Malfa 153, Palermo, Italy
| | - Annalisa Pinsino
- Istituto per la Ricerca e l'Innovazione Biomedica-Consiglio Nazionale delle Ricerche, Via Ugo la Malfa 153, Palermo, Italy
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università di Palermo, Via Archirafi 18, Palermo, Italy.
| |
Collapse
|
10
|
Yoshihisa Y, Andoh T, Rehman MU, Shimizu T. The regulation of protein kinase casein kinase II by apigenin is involved in the inhibition of ultraviolet B-induced macrophage migration inhibitory factor-mediated hyperpigmentation. Phytother Res 2019; 34:1320-1328. [PMID: 31840901 DOI: 10.1002/ptr.6597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 11/08/2022]
Abstract
Ultraviolet (UV) radiation elicits melanogenesis and pigmentation in the skin. Apigenin (4',5,7-trihydroxyflavone [AGN]) is a plant flavone contained in various herbs, fruits, and vegetables. We herein investigated antimelanogenic properties of AGN and the molecular mechanisms of the action of AGN. In UVB-treated mice, AGN inhibited cutaneous hyperpigmentation and macrophage migration inhibitory factor (MIF) expression as a melanogenesis-related key factor. In mouse keratinocytes, AGN inhibited the expression of MIF and also the related factors (e.g., stem cell factor and proteinase-activated receptor 2) induced by MIF. In addition to ellagic acid as a casein kinase II (CK2) inhibitor, AGN suppressed CK2 enzymatic activity and UVB-induced CK2 expression and subsequent phosphorylation of IκB and MIF expression. These results suggest that AGN inhibits UVB-induced hyperpigmentation through the regulation of CK2-mediated MIF expression in keratinocytes.
Collapse
Affiliation(s)
- Yoko Yoshihisa
- Department of Dermatology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tsugunobu Andoh
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Mati Ur Rehman
- Department of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Tadamichi Shimizu
- Department of Dermatology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
11
|
Mishra A, Sundaravadivel P, Tripathi SK, Jha RK, Badrukhiya J, Basak N, Anerao I, Sharma A, Idowu AE, Mishra A, Pandey S, Kumar U, Singh S, Nizamuddin S, Tupperwar NC, Jha AN, Thangaraj K. Variations in macrophage migration inhibitory factor gene are not associated with visceral leishmaniasis in India. J Infect Public Health 2019; 12:380-387. [PMID: 30611734 DOI: 10.1016/j.jiph.2018.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/24/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The host genetic factors play important role in determining the outcome of visceral leishmaniasis (VL). Macrophage migration inhibitory factor (MIF) is an important host cytokine, which is a key regulator of innate immune system. Genetic variants in MIF gene have been found to be associated with several inflammatory and infectious diseases. Role of MIF is well documented in leishmaniasis diseases, including Indian visceral leishmaniasis, where elevated level of serum MIF has been associated with VL phenotypes. However, there was no genetic study to correlate MIF variants in VL, therefore, we aimed to study the possible association of three reported MIF gene variants -794 CATT, -173G > C and non-coding RNA gene LOC284889 in Indian VL phenotype. METHODS Study subjects comprised of 214 VL patients along with ethnically and demographically matched 220 controls from VL endemic regions of Bihar state in India. RESULTS We found no significant difference between cases and controls in allelic, genotypic and haplotype frequency of the markers analysed [-794 CATT repeats (χ2=0.86; p=0.35; OR=0.85; 95% CI=0.61-1.19); -173 G>C polymorphism (χ2=1.11; p=0.29; OR=0.83; 95% CI=0.59-1.16); and LOC284889 (χ2=0.78; p=0.37; OR=0.86; 95% CI=0.61-1.20)]. CONCLUSION Since we did not find any significant differences between case and control groups, we conclude that sequencing of complete MIF gene and extensive study on innate and adaptive immunity genes may help in identifying genetic variations that are associated with VL susceptibility/resistance among Indians.
Collapse
Affiliation(s)
- Anshuman Mishra
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India; Vinoba Bhave Research Institute, Allahabad, India; Institute of Advanced Materials, Linkoping, Sweden
| | | | | | - Rajan Kumar Jha
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Nipa Basak
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India; Academy of Scientific and Innovative Research, India
| | - Isha Anerao
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Akshay Sharma
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Ajayi Ebenezer Idowu
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India; Osun State University, Oshogbo, Nigeria
| | | | | | - Umesh Kumar
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Sakshi Singh
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | | - Aditya Nath Jha
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India; Sickle Cell Institute Chhattisgarh, Raipur, India
| | | |
Collapse
|
12
|
Li J, Tang Y, Tang PMK, Lv J, Huang XR, Carlsson-Skwirut C, Da Costa L, Aspesi A, Fröhlich S, Szczęśniak P, Lacher P, Klug J, Meinhardt A, Fingerle-Rowson G, Gong R, Zheng Z, Xu A, Lan HY. Blocking Macrophage Migration Inhibitory Factor Protects Against Cisplatin-Induced Acute Kidney Injury in Mice. Mol Ther 2018; 26:2523-2532. [PMID: 30077612 DOI: 10.1016/j.ymthe.2018.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/08/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is elevated in patients with acute kidney injury (AKI) and is suggested as a potential predictor for renal replacement therapy in AKI. In this study, we found that MIF also plays a pathogenic role and is a therapeutic target for AKI. In a cisplatin-induced AKI mouse model, elevated plasma MIF correlated with increased serum creatinine and the severity of renal inflammation and tubular necrosis, whereas deletion of MIF protected the kidney from cisplatin-induced AKI by largely improving renal functional and histological injury, and suppressing renal inflammation including upregulation of cytokines such as interleukin (IL)-1β, tumor necrosis factor-alpha (TNF-α), IL-6, inducible nitric oxide synthase (iNOS), MCP-1, IL-8, and infiltration of macrophages, neutrophils, and T cells. We next developed a novel therapeutic strategy for AKI by blocking the endogenous MIF with an MIF inhibitor, ribosomal protein S19 (RPS19). Similar to the MIF-knockout mice, treatment with RPS19, but not the mutant RPS19, suppressed cisplatin-induced AKI. Mechanistically, we found that both genetic knockout and pharmacological inhibition of MIF protected against AKI by inactivating the CD74-nuclear factor κB (NF-κB) signaling. In conclusion, MIF is pathogenic in cisplatin-induced AKI. Targeting MIF with an MIF inhibitor RPS19 could be a promising therapeutic potential for AKI.
Collapse
Affiliation(s)
- Jinhong Li
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ying Tang
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Patrick M K Tang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Lv
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Christine Carlsson-Skwirut
- Department of Woman and Child Health, Paediatric Endocrinology Unit, Astrid Lindgren Children's Hospital, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Lydie Da Costa
- AP-HP, Service d'Hématologie Biologique, Hôpital R. Debré, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; INSERM U1149, CRI, Faculté de Médecine Bichat-Claude Bernard, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Anna Aspesi
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy
| | - Suada Fröhlich
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Pawel Szczęśniak
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Philipp Lacher
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Jörg Klug
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Günter Fingerle-Rowson
- Department I of Internal Medicine, University Hospital Cologne and Center for Integrated Oncology Köln-Bonn, Cologne, Germany
| | - Rujun Gong
- Division of Kidney Diseases and Hypertension, Rhode Island Hospital, Brown University School of Medicine, Providence, RI, USA
| | - Zhihua Zheng
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Anping Xu
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Department of Anatomical and Cellular Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
13
|
Parekh A, Das S, Parida S, Das CK, Dutta D, Mallick SK, Wu PH, Kumar BNP, Bharti R, Dey G, Banerjee K, Rajput S, Bharadwaj D, Pal I, Dey KK, Rajesh Y, Jena BC, Biswas A, Banik P, Pradhan AK, Das SK, Das AK, Dhara S, Fisher PB, Wirtz D, Mills GB, Mandal M. Multi-nucleated cells use ROS to induce breast cancer chemo-resistance in vitro and in vivo. Oncogene 2018; 37:4546-4561. [PMID: 29743594 DOI: 10.1038/s41388-018-0272-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/23/2018] [Accepted: 02/09/2018] [Indexed: 11/09/2022]
Abstract
Although there is a strong correlation between multinucleated cells (MNCs) and cancer chemo-resistance in variety of cancers, our understanding of how multinucleated cells modulate the tumor micro-environment is limited. We captured multinucleated cells from triple-negative chemo-resistant breast cancers cells in a time frame, where they do not proliferate but rather significantly regulate their micro-environment. We show that oxidatively stressed MNCs induce chemo-resistance in vitro and in vivo by secreting VEGF and MIF. These factors act through the RAS/MAPK pathway to induce chemo-resistance by upregulating anti-apoptotic proteins. In MNCs, elevated reactive oxygen species (ROS) stabilizes HIF-1α contributing to increase production of VEGF and MIF. Together the data indicate, that the ROS-HIF-1α signaling axis is very crucial in regulation of chemo-resistance by MNCs. Targeting ROS-HIF-1α in future may help to abrogate drug resistance in breast cancer.
Collapse
Affiliation(s)
- Aditya Parekh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Sheetal Parida
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Chandan Kanta Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Debabrata Dutta
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Sanjaya K Mallick
- BD Biosciences-Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata, West Bengal, India
| | - Pei-Hsun Wu
- Department of chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - B N Prashanth Kumar
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Rashmi Bharti
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Goutam Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Kacoli Banerjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Shashi Rajput
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Deblina Bharadwaj
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Ipsita Pal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Kaushik Kumar Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Yetirajam Rajesh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Bikash Chandra Jena
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Payel Banik
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Santanu Dhara
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Paul B Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Denis Wirtz
- Department of chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Gordon B Mills
- Department of Systems Biology, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
14
|
Identification of CPE and GAIT elements in 3'UTR of macrophage migration inhibitory factor (MIF) involved in inflammatory response induced by LPS in Ciona robusta. Mol Immunol 2018; 99:66-74. [PMID: 29702356 DOI: 10.1016/j.molimm.2018.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/14/2018] [Accepted: 04/17/2018] [Indexed: 01/06/2023]
Abstract
Innate immune responses face infectious microorganisms by inducing inflammatory responses. Multiple genes within distinct functional categories are coordinately and temporally regulated by transcriptional 'on' and 'off' switches that account for the specificity of gene expression in response to external stimuli. Mechanisms that control transcriptional and post-transcriptional regulation are important in coordinating the initiation and resolution of inflammation. Macrophage migration inhibitory factor (MIF) is an important cytokine that, in Ciona robusta, is related to inflammatory response. It is well known that in C. robusta, formerly known as Ciona intestinalis, the pharynx is involved in the inflammatory reaction induced by lipopolysaccharide (LPS) injection in the body wall. Using this biological system, we describe the identification of two C. robusta MIFs (CrMIF1 and CrMIF2). The phylogenetic tree and modeling support a close relationship with vertebrate MIF family members. CrMIF1 and CrMIF2 possess two evolutionally conserved catalytic sites: a tautomerase and an oxidoreductase site with a conserved CXXC motif. Real-time PCR analysis shows a prompt expression induced by LPS inoculation in CrMIF1 and a late upregulation of CrMIF2 and in silico analyses of 3'UTR show a cis-acting GAIT element and a CPE element in 3'-UTR, which are not present in the 3'-UTR of CrMIF1, suggesting that different transcriptional and post-transcriptional control mechanisms are involved in the regulation of gene expression of MIF during inflammatory response in C. robusta.
Collapse
|
15
|
Macrophage migration inhibitory factor activates inflammatory responses of astrocytes through interaction with CD74 receptor. Oncotarget 2018; 8:2719-2730. [PMID: 27926507 PMCID: PMC5356836 DOI: 10.18632/oncotarget.13739] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/22/2016] [Indexed: 01/17/2023] Open
Abstract
Astrocytes, the major glial cell population of the central nervous system (CNS), play important physiological roles related to CNS homeostasis. Growing evidence demonstrates that astrocytes trigger innate immune responses under challenge of a variety of proinflammatory cytokines. Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine mainly secreted from monocytes/macrophages, is involved in inflammation-associated pathophysiology. Here, we displayed that expression of MIF significantly increased following spinal cord injury, in colocalization with microglia and astrocytes. MIF elicited inflammatory responses of astrocytes via activation of CD74 receptor and extracellular signal-related kinase (ERK) pathway. Transcriptome analysis revealed that inflammation-related factors cholesterol 25-hydroxylase (Ch25h) and phospholipase A2-IIA (Pla2g2a), downstream of MIF/CD74 axis, were potentially implicated in the mediating inflammatory response of astrocytes. Our results provided a new target for interference of CNS inflammation after insults.
Collapse
|
16
|
Sparkes A, De Baetselier P, Roelants K, De Trez C, Magez S, Van Ginderachter JA, Raes G, Bucala R, Stijlemans B. Reprint of: The non-mammalian MIF superfamily. Immunobiology 2017; 222:858-867. [PMID: 28552269 DOI: 10.1016/j.imbio.2017.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/03/2016] [Accepted: 10/10/2016] [Indexed: 01/31/2023]
Abstract
Macrophage migration inhibitory factor (MIF) was first described as a cytokine 50 years ago, and emerged in mammals as a pleiotropic protein with pro-inflammatory, chemotactic, and growth-promoting activities. In addition, MIF has gained substantial attention as a pivotal upstream mediator of innate and adaptive immune responses and with pathologic roles in several diseases. Of less importance in mammals is an intrinsic but non-physiologic enzymatic activity that points to MIF's evolution from an ancient defense molecule. Therefore, it is not surprising that mif-like genes also have been found across a range of different organisms including bacteria, plants, protozoa, helminths, molluscs, arthropods, fish, amphibians and birds. While Genebank analysis identifying mif-like genes across species is extensive, contained herein is an overview of the non-mammalian MIF-like proteins that have been most well studied experimentally. For many of these organisms, MIF contributes to an innate defense system or plays a role in development. For parasitic organisms however, MIF appears to function as a virulence factor aiding in the establishment or persistence of infection by modulating the host immune response. Consequently, a combined targeting of both parasitic and host MIF could lead to more effective treatment strategies for parasitic diseases of socioeconomic importance.
Collapse
Affiliation(s)
- Amanda Sparkes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Patrick De Baetselier
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Kim Roelants
- Amphibian Evolution Lab, Department of Biology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Carl De Trez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB Structural Biology Research Center, Brussels, Belgium
| | - Stefan Magez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB Structural Biology Research Center, Brussels, Belgium; Laboratory for Biomedical Research, Ghent University Global Campus, Yeonsu-Gu, Incheon, South Korea
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
| |
Collapse
|
17
|
Sparkes A, De Baetselier P, Roelants K, De Trez C, Magez S, Van Ginderachter JA, Raes G, Bucala R, Stijlemans B. The non-mammalian MIF superfamily. Immunobiology 2017; 222:473-482. [PMID: 27780588 PMCID: PMC5293613 DOI: 10.1016/j.imbio.2016.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/03/2016] [Accepted: 10/10/2016] [Indexed: 01/09/2023]
Abstract
Macrophage migration inhibitory factor (MIF) was first described as a cytokine 50 years ago, and emerged in mammals as a pleiotropic protein with pro-inflammatory, chemotactic, and growth-promoting activities. In addition, MIF has gained substantial attention as a pivotal upstream mediator of innate and adaptive immune responses and with pathologic roles in several diseases. Of less importance in mammals is an intrinsic but non-physiologic enzymatic activity that points to MIF's evolution from an ancient defense molecule. Therefore, it is not surprising that mif-like genes also have been found across a range of different organisms including bacteria, plants, protozoa, helminths, molluscs, arthropods, fish, amphibians and birds. While Genebank analysis identifying mif-like genes across species is extensive, contained herein is an overview of the non-mammalian MIF-like proteins that have been most well studied experimentally. For many of these organisms, MIF contributes to an innate defense system or plays a role in development. For parasitic organisms however, MIF appears to function as a virulence factor aiding in the establishment or persistence of infection by modulating the host immune response. Consequently, a combined targeting of both parasitic and host MIF could lead to more effective treatment strategies for parasitic diseases of socioeconomic importance.
Collapse
Affiliation(s)
- Amanda Sparkes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium
| | - Patrick De Baetselier
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium
| | - Kim Roelants
- Amphibian Evolution Lab, Department of Biology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Carl De Trez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB Structural Biology Research Center, Brussels, Belgium
| | - Stefan Magez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB Structural Biology Research Center, Brussels, Belgium; Laboratory for Biomedical Research, Ghent University Global Campus, Yeonsu-Gu, Incheon, South Korea
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium.
| |
Collapse
|
18
|
Kim KW, Kim HR. Macrophage migration inhibitory factor: a potential therapeutic target for rheumatoid arthritis. Korean J Intern Med 2016; 31:634-42. [PMID: 27169879 PMCID: PMC4939511 DOI: 10.3904/kjim.2016.098] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/26/2016] [Indexed: 12/27/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is originally identified in the culture medium of activated T lymphocytes as a soluble factor that inhibits the random migration of macrophages. MIF is now recognized as a multipotent cytokine involved in the regulation of immune and inf lammatory responses. In rheumatoid arthritis (RA), MIF promotes inf lammatory responses by inducing proinflammatory cytokines and tissue-degrading molecules, promoting the proliferation and survival of synovial fibroblasts, stimulating neutrophil chemotaxis, and regulating angiogenesis and osteoclast differentiation. Expression of MIF in synovial tissue and synovial fluid levels of MIF are elevated in RA patients. Specifically, MIF levels correlate with RA disease activity and high levels are associated with bone erosion. In animal models of RA, the genetic and therapeutic inhibition of MIF has been shown to control inflammation and bone destruction. Based on the role of MIF in RA pathogenesis, small molecular inhibitors targeting it or its receptor pathways could provide a new therapeutic option for RA patients.
Collapse
Affiliation(s)
- Kyoung-Woon Kim
- Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
- Correspondence to Hae-Rim Kim, M.D. Division of Rheumatology, Department of Internal Medicine, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea Tel: +82-2-2030-7542 Fax: +82-2-2030-7748 E-mail:
| |
Collapse
|
19
|
Bay-Richter C, Janelidze S, Sauro A, Bucala R, Lipton J, Deierborg T, Brundin L. Behavioural and neurobiological consequences of macrophage migration inhibitory factor gene deletion in mice. J Neuroinflammation 2015; 12:163. [PMID: 26338025 PMCID: PMC4558780 DOI: 10.1186/s12974-015-0387-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 08/24/2015] [Indexed: 01/15/2023] Open
Abstract
Background Evidence from clinical studies and animal models show that inflammation can lead to the development of depression. Macrophage migration inhibitory factor (MIF) is an important multifunctional cytokine that is synthesized by several cell types in the brain. MIF can increase production of other cytokines, activates cyclooxygenase (COX)-2 and can counter-regulate anti-inflammatory effects of glucocorticoids. Increased plasma levels of MIF are associated with hypothalamic–pituitary–adrenal (HPA) axis dysregulation and depressive symptoms in patients. In contrast, MIF knockout (KO) mice have been found to exhibit increased depressive-like behaviour. The exact role for MIF in depression is therefore still controversial. To further understand the role of MIF in depression, we studied depressive-like behaviour in congenic male and female MIF KO mice and wild-type (WT) littermates and the associated neurobiological mechanisms underlying the behavioural outcome. Methods MIF KO and WT mice were tested for spontaneous locomotor activity in the open-field test, anhedonia-like behaviour in the sucrose preference test (SPT), as well as behavioural despair in the forced swim test (FST) and tail suspension test (TST). Brain and serum levels of cytokines, the enzymes COX-2 and indoleamine-2,3-dioxygenase (IDO) and the glucocorticoid hormone corticosterone were measured by RT-qPCR and/or high-sensitivity electrochemiluminescence-based multiplex immunoassays. Monoamines and metabolites were examined using HPLC. Results We found that MIF KO mice of both sexes displayed decreased depressive-like behaviour as measured in the FST. In the TST, a similar, but non-significant, trend was also found. IFN-γ levels were decreased, and dopamine metabolism increased in MIF KO mice. Decreased brain IFN-γ levels predicted higher striatal dopamine levels, and high dopamine levels in turn were associated with reduced depressive-like behaviour. In the SPT, there was a sex-specific discrepancy, where male MIF KO mice showed reduced anhedonia-like behaviour whereas female KO mice displayed increased anhedonia-like behaviour. Our results suggest that this relates to the increased corticosterone levels detected in female, but not male, MIF KO mice. Conclusions Our findings support that MIF is involved in the generation of depressive-like symptoms, potentially by the effects of IFN-γ on dopamine metabolism. Our data further suggests a sex-specific regulation of the involved mechanisms.
Collapse
Affiliation(s)
- Cecilie Bay-Richter
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark.
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | - Analise Sauro
- Department of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, MI, USA.
| | - Richard Bucala
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA.
| | - Jack Lipton
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA.
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, BMC, Lund University, Lund, Sweden.
| | - Lena Brundin
- Department of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, MI, USA. .,Laboratory of Behavioral Medicine, Van Andel Research Institute, Grand Rapids, MI, USA.
| |
Collapse
|
20
|
A Secreted MIF Cytokine Enables Aphid Feeding and Represses Plant Immune Responses. Curr Biol 2015; 25:1898-903. [PMID: 26119751 DOI: 10.1016/j.cub.2015.05.047] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/23/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022]
Abstract
Aphids attack virtually all plant species and cause serious crop damages in agriculture. Despite their dramatic impact on food production, little is known about the molecular processes that allow aphids to exploit their host plants. To date, few aphid salivary proteins have been identified that are essential for aphid feeding, and their nature and function remain largely unknown. Here, we show that a macrophage migration inhibitory factor (MIF) is secreted in aphid saliva. In vertebrates, MIFs are important pro-inflammatory cytokines regulating immune responses. MIF proteins are also secreted by parasites of vertebrates, including nematodes, ticks, and protozoa, and participate in the modulation of host immune responses. The finding that a plant parasite secretes a MIF protein prompted us to question the role of the cytokine in the plant-aphid interaction. We show here that expression of MIF genes is crucial for aphid survival, fecundity, and feeding on a host plant. The ectopic expression of aphid MIFs in leaf tissues inhibits major plant immune responses, such as the expression of defense-related genes, callose deposition, and hypersensitive cell death. Functional complementation analyses in vivo allowed demonstrating that MIF1 is the member of the MIF protein family that allows aphids to exploit their host plants. To our knowledge, this is the first report of a cytokine that is secreted by a parasite to modulate plant immune responses. Our findings suggest a so-far unsuspected conservation of infection strategies among parasites of animal and plant species.
Collapse
|
21
|
Lerch JK, Puga DA, Bloom O, Popovich PG. Glucocorticoids and macrophage migration inhibitory factor (MIF) are neuroendocrine modulators of inflammation and neuropathic pain after spinal cord injury. Semin Immunol 2014; 26:409-14. [DOI: 10.1016/j.smim.2014.03.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 11/29/2022]
|
22
|
Zhang X, Chen L, Wang Y, Ding Y, Peng Z, Duan L, Ju G, Ren Y, Wang X. Macrophage migration inhibitory factor promotes proliferation and neuronal differentiation of neural stem/precursor cells through Wnt/β-catenin signal pathway. Int J Biol Sci 2013; 9:1108-20. [PMID: 24339732 PMCID: PMC3858584 DOI: 10.7150/ijbs.7232] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/23/2013] [Indexed: 01/12/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a highly conserved and evolutionarily ancient mediator with pleiotropic effects. Recent studies demonstrated that the receptors of MIF, including CD44, CXCR2, CXCR4 and CD74, are expressed in the neural stem/progenitor cells (NSPCs). The potential regulatory effect of MIF on NSPCs proliferation and neuronal differentiation, however, is largely unknown. Here, we investigated the effect of MIF on NSPC proliferation and neuronal differentiation, and further examined the signal pathway by which MIF transduced these signal effects in mouse NSPCs in vitro. The results showed that both Ki67-positive cells and neurosphere volumes were increased in a dose-dependent manner following MIF treatment. Furthermore, the expression of nuclear β-catenin was significantly stronger in MIF-stimulated groups than that in control groups. Conversely, administration of IWR-1, the inhibitor of Wnt/β-catenin pathway, significantly inhibited the proliferative effect of MIF on NSPCs. Immunostaining and Western blot further indicated that doublecortin (DCX) and Tuj 1, two neuronal markers, were evidently increased with MIF stimulation during NSPC differentiation, and there were more Tuj1-positive cells migrated out from neurospheres in MIF-stimulated groups than those in control groups. During NSPC differentiation, MIF increased the activity of β-galactosidase that responds to Wnt/β-catenin signaling. Wnt1 and β-catenin proteins were also up-regulated with MIF stimulation. Moreover, the expression of DCX and Tuj 1 was inhibited significantly by IWR-1. Taken together, the present study indicated that MIF enhances NSPC proliferation and promotes the neuronal differentiation, by activating Wnt/β-catenin signal pathway. The interaction between MIF and Wnt/β-catenin signal pathway may play an important role in modulating NSPC renewal and fate during brain development.
Collapse
Affiliation(s)
- Xijing Zhang
- 1. Department of Anesthesiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wasiel AA, Baas BJ, Zandvoort E, Quax WJ, Poelarends GJ. Dehalogenation of an Anthropogenic Compound by an Engineered Variant of the Mouse Cytokine Macrophage Migration Inhibitory Factor. Chembiochem 2012; 13:1270-3. [DOI: 10.1002/cbic.201200153] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Indexed: 11/12/2022]
|
24
|
Lue H, Dewor M, Leng L, Bucala R, Bernhagen J. Activation of the JNK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on CXCR4 and CD74. Cell Signal 2011; 23:135-44. [PMID: 20807568 PMCID: PMC3586206 DOI: 10.1016/j.cellsig.2010.08.013] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 08/17/2010] [Accepted: 08/24/2010] [Indexed: 12/31/2022]
Abstract
c-Jun N-terminal kinase (JNK) is a member of the mitogen-activated protein kinase (MAPK) family and controls essential processes such as inflammation, cell differentiation, and apoptosis. JNK signalling is triggered by extracellular signals such as cytokines and environmental stresses. Macrophage migration inhibitory factor (MIF) is a pleiotropic pro-inflammatory cytokine with chemokine-like functions in leukocyte recruitment and atherosclerosis. MIF promotes MAPK signalling through ERK1/2, while it can either activate or inhibit JNK phosphorylation, depending on the cell type and underlying stimulation context. MIF activities are mediated by non-cognate interactions with the CXC chemokine receptors CXCR2 and CXCR4 or by ligation of CD74, which is the cell surface expressed form of the class II invariant chain. ERK1/2 signalling stimulated by MIF is dependent on CD74, but the receptor pathway involved in MIF activation of the JNK pathway is unknown. Here we comprehensively characterize the stimulatory effect of MIF on the canonical JNK/c-Jun/AP-1 pathway in fibroblasts and T cell lines and identify the upstream signalling components. Physiological concentrations of recombinant MIF triggered the phosphorylation of JNK and c-Jun and rapidly activated AP-1. In T cells, MIF-mediated activation of the JNK pathway led to upregulated gene expression of the inflammatory chemokine CXCL8. Activation of JNK signalling by MIF involved the upstream kinases PI3K and SRC and was found to be dependent on CXCR4 and CD74. Together, these data show that the CXCR4/CD74/SRC/PI3K axis mediates a rapid and transient activation of the JNK pathway as triggered by the inflammatory cytokine MIF in T cells and fibroblasts.
Collapse
Affiliation(s)
- Hongqi Lue
- Department of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Manfred Dewor
- Department of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | - Lin Leng
- Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520-8031, USA
| | - Richard Bucala
- Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520-8031, USA
| | - Jürgen Bernhagen
- Department of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| |
Collapse
|
25
|
Wasiel AA, Rozeboom HJ, Hauke D, Baas BJ, Zandvoort E, Quax WJ, Thunnissen AMWH, Poelarends GJ. Structural and functional characterization of a macrophage migration inhibitory factor homologue from the marine cyanobacterium Prochlorococcus marinus . Biochemistry 2010; 49:7572-81. [PMID: 20715791 DOI: 10.1021/bi1008276] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional mammalian cytokine, which exhibits tautomerase and oxidoreductase activity. MIF homologues with pairwise sequence identities to human MIF ranging from 31% to 41% have been detected in various cyanobacteria. The gene encoding the MIF homologue from the marine cyanobacterium Prochlorococcus marinus strain MIT9313 has been cloned and the corresponding protein (PmMIF) overproduced, purified, and subjected to functional and structural characterization. Kinetic and (1)H NMR spectroscopic studies show that PmMIF tautomerizes phenylenolpyruvate and (p-hydroxyphenyl)enolpyruvate at low levels. The N-terminal proline of PmMIF is critical for these reactions because the P1A mutant has strongly reduced tautomerase activities. PmMIF shows high structural homology with mammalian MIFs as revealed by a crystal structure of PmMIF at 1.63 A resolution. MIF contains a Cys-X-X-Cys motif that mediates oxidoreductase activity, which is lacking from PmMIF. Engineering of the motif into PmMIF did not result in oxidoreductase activity but increased the tautomerase activity 8-fold. The shared tautomerase activities and the conservation of the beta-alpha-beta structural fold and key functional groups suggest that eukaryotic MIFs and cyanobacterial PmMIF are related by divergent evolution from a common ancestor. While several MIF homologues have been identified in eukaryotic parasites, where they are thought to play a role in modulating the host immune response, PmMIF is the first nonparasitic, bacterial MIF-like protein characterized in detail. This work sets the stage for future studies which could address the question whether a MIF-like protein from a free-living bacterium possesses immunostimulatory features similar to those of mammalian MIFs and MIF-like proteins found in parasitic nematodes and protozoa.
Collapse
Affiliation(s)
- Anna A Wasiel
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Hare AA, Leng L, Gandavadi S, Du X, Cournia Z, Bucala R, Jorgensen WL. Optimization of N-benzyl-benzoxazol-2-ones as receptor antagonists of macrophage migration inhibitory factor (MIF). Bioorg Med Chem Lett 2010; 20:5811-4. [PMID: 20728358 PMCID: PMC2939296 DOI: 10.1016/j.bmcl.2010.07.129] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/28/2010] [Accepted: 07/29/2010] [Indexed: 11/20/2022]
Abstract
The cytokine MIF is involved in inflammation and cell proliferation via pathways initiated by its binding to the transmembrane receptor CD74. MIF also exhibits keto-enol tautomerase activity, believed to be vestigial in mammals. Starting from a 1 μM hit from virtual screening, substituted benzoxazol-2-ones have been discovered as antagonists with IC(50) values as low as 7.5 nM in a tautomerase assay and 80 nM in a MIF-CD74 binding assay. Additional studies for one of the potent inhibitors demonstrated that it is not a covalent inhibitor of MIF and that it attenuates MIF-dependent ERK1/2 phosphorylation in human synovial fibroblasts.
Collapse
Affiliation(s)
- Alissa A. Hare
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, USA
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520-8066, USA
| | | | - Xin Du
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520-8066, USA
| | - Zoe Cournia
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, USA
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520-8066, USA
| | | |
Collapse
|
27
|
Jorgensen WL, Gandavadi S, Du X, Hare AA, Trofimov A, Leng L, Bucala R. Receptor agonists of macrophage migration inhibitory factor. Bioorg Med Chem Lett 2010; 20:7033-6. [PMID: 20971005 DOI: 10.1016/j.bmcl.2010.09.118] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/21/2010] [Accepted: 09/23/2010] [Indexed: 11/25/2022]
Abstract
The cytokine MIF is involved in inflammation and cell proliferation via pathways initiated by its binding to the transmembrane receptor CD74. MIF also promotes AMPK activation with potential benefits for response to myocardial infarction and ischemia-reperfusion. Structure-based molecular design has led to the discovery of not only antagonists, but also the first agonists of MIF-CD74 binding. The compounds contain a triazole core that is readily assembled via Cu-catalyzed click chemistry. The agonist and antagonist behaviors were confirmed via study of MIF-dependent ERK1/2 phosphorylation in human fibroblasts.
Collapse
|
28
|
Pettorini BL, Frassanito P, Caldarelli M, Tamburrini G, Massimi L, Di Rocco C. Molecular pathogenesis of craniopharyngioma: switching from a surgical approach to a biological one. Neurosurg Focus 2010; 28:E1. [DOI: 10.3171/2010.1.focus09300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Craniopharyngioma has long been considered a benign tumor because of its pathological aspect. This primordial view of craniopharyngioma fit with the primitive treatment attempts based on blind resection of the tumor each time it recurred. The limits of this management strategy were proven early by the high morbidity related to the resection and recurrence risk despite radical lesion removal. Nowadays, craniopharyngioma must be considered a complex molecular disease, and a detailed explanation of the mechanisms underlying its aggressive biological and clinical behavior, despite some benign pathological features, would be the first step toward defining the best management of craniopharyngioma. Indeed, advances in the knowledge of the molecular mechanisms at the base of craniopharyngioma oncogenesis will lead to comprehension of the critical checkpoints involved in neoplastic transformation. The final research target will be the definition of new biological agents able to reverse the neoplastic process by acting on these critical checkpoints. This biological approach will lead to a refined therapy combining higher efficacy and safety with lower morbidity. In this paper the authors reveal state-of-the-art comprehension of the molecular biology of craniopharyngioma and the consequent therapeutic implications.
Collapse
|
29
|
Harrison RA, Sumners C. Redox regulation of macrophage migration inhibitory factor expression in rat neurons. Biochem Biophys Res Commun 2009; 390:171-5. [PMID: 19799867 DOI: 10.1016/j.bbrc.2009.09.112] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 09/25/2009] [Indexed: 01/29/2023]
Abstract
Macrophage migration inhibitory factor (MIF) expression is induced by angiotensin II (Ang II) in normal rat neurons and serves a negative regulatory role by blunting the chronotropic actions of this peptide. The aim here was to determine whether hydrogen peroxide (H(2)O(2)), a reactive oxygen species (ROS) that is a key intracellular mediator of the neuronal actions of Ang II, is a trigger for MIF production in neurons. Thus, we tested the effects of H(2)O(2) on MIF expression in primary neuronal cultures from newborn normotensive (Wistar Kyoto [WKY] or Sprague-Dawley [SD]) rat brain, cells that respond to Ang II by increasing MIF levels. Treatment of WKY or SD rat neuronal cultures with a non-cytotoxic concentration of H(2)O(2) elicited a significant, time-dependent increase in MIF mRNA and protein levels. Glucose oxidase, which produces H(2)O(2) via oxidation of glucose in the cell-culture medium, elicited a similar increase in neuronal MIF mRNA levels. The stimulatory action of H(2)O(2) was not apparent in neuronal cultures from spontaneously hypertensive rats (SHR), cells that fail to express increased MIF in response to Ang II. Finally, preincubation of SD rat cultures with either polyethylene glycol-catalase or actinomycin D abolished the H(2)O(2)-induced increase in MIF, suggesting that this ROS is acting intracellularly to increase transcription of the MIF gene. These results suggest the presence of a redox regulatory mechanism for induction of MIF in normotensive rat neurons.
Collapse
Affiliation(s)
- Rachael A Harrison
- Department of Physiology and Functional Genomics & McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA
| | | |
Collapse
|
30
|
Activation of nuclear factor kappa B and induction of migration inhibitory factor in tumors by surgical stress of laparotomy versus carbon dioxide pneumoperitoneum: an animal experiment. Surg Endosc 2009; 24:578-83. [PMID: 19609608 DOI: 10.1007/s00464-009-0609-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Revised: 05/08/2009] [Accepted: 06/16/2009] [Indexed: 12/23/2022]
Abstract
BACKGROUND Surgical trauma may be associated with enhanced tumor growth and establishment. The authors studied the effect of carbon dioxide (CO(2)) pneumoperitoneum versus laparotomy on tumor necrosis factor-alpha (TNFalpha), migration inhibitory factor (MIF) expression, and nuclear factor kappa B (NFkappaB) activity in human gastric cancer. METHODS Nude mice were inoculated intraperitoneally with human gastric cancer cells (MKN45). Then laparotomy, CO(2) pneumoperitoneum, and anesthesia alone were performed randomly. Tumor growth and associated TNFalpha and MIF expression and NFkappaB activity were determined. RESULTS Total tumor weight, especially at the anterior abdominal wall, was higher after laparotomy than after CO(2) pneumoperitoneum (p < 0.05). The mRNA expression of TNFalpha was higher 24 and 48 h after laparotomy than after CO(2) pneumoperitoneum (p < 0.05 and p < 0.01, respectively). At all the examined time points, MIF mRNA expression also was higher after laparotomy than after CO(2) pneumoperitoneum (p < 0.05 until 1 week or p < 0.01 at 2 weeks). The NFkappaB protein was more activated after laparotomy than after CO(2) pneumoperitoneum 6 h subsequent to surgical procedures. CONCLUSION After CO(2) pneumoperitoneum, tumors have less TNFalpha and MIF expression and less NFkappaB activity than after laparotomy. This may be associated with less tumor growth, supporting minimal invasive techniques in gastrointestinal oncologic surgery.
Collapse
|
31
|
Prieto-Lafuente L, Gregory WF, Allen JE, Maizels RM. MIF homologues from a filarial nematode parasite synergize with IL-4 to induce alternative activation of host macrophages. J Leukoc Biol 2009; 85:844-54. [PMID: 19179453 PMCID: PMC2691607 DOI: 10.1189/jlb.0808459] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 12/19/2008] [Accepted: 12/22/2008] [Indexed: 12/18/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a highly conserved cytokine considered to exert wide-ranging, proinflammatory effects on the immune system. Recently, members of this gene family have been discovered in a number of invertebrate species, including parasitic helminths. However, chronic helminth infections are typically associated with a Th2-dominated, counter-inflammatory phenotype, in which alternatively activated macrophages (AAMs) are prominent. To resolve this apparent paradox, we have analyzed the activity of two helminth MIF homologues from the filarial nematode Brugia malayi, in comparison with the canonical MIF from the mouse. We report that murine MIF (mMIF) and Brugia MIF proteins induce broadly similar effects on bone marrow-derived mouse macrophages, eliciting a measured release of proinflammatory cytokines. In parallel, MIF was found to induce up-regulation of IL-4R on macrophages, which when treated in vitro with MIF in combination with IL-4, expressed markers of alternative activation [arginase, resistin-like molecule alpha (RELM-alpha) or found in inflammatory zone 1, Ym-1, murine macrophage mannose receptor] and differentiated into functional AAMs with in vitro-suppressive ability. Consistent with this finding, repeated in vivo administration of Brugia MIF induced expression of alternative macrophage activation markers. As mMIF did not induce RELM-alpha or Ym-1 in vivo, alternative activation may require components of the adaptive immune response to Brugia MIF, such as the production of IL-4. Hence, MIF may accentuate macrophage activation according to the polarity of the environment, thus promoting AAM differentiation in the presence of IL-4-inducing parasitic helminths.
Collapse
Affiliation(s)
- Lidia Prieto-Lafuente
- Institute of Immunology and Infection Research, University of Edinburgh, West Mains Road, Edinburgh EH9 3JT, UK
| | | | | | | |
Collapse
|
32
|
Zhang C, Hou G, Liang T, Song J, Qu L, Chen F, Xu J, Wang D, Han J. A prospective study of macrophage migration inhibitory factor as a marker of inflammatory detection. J Cell Mol Med 2008; 13:4077-83. [PMID: 19602037 PMCID: PMC4516554 DOI: 10.1111/j.1582-4934.2008.00537.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study was to evaluate whether macrophage migration inhibitory factor (MIF) can be used as a better marker of inflammatory detection through the biodistribution and inflammatory imaging study with 131I-labelled anti-MIF McAb and control antibody in inflammatory model mice. The mRNA and protein expression of MIF in inflammatory lesions were proved by RT-PCR and immunohistochemistry. The model mice were injected with 3.7 MBq of each agent and killed at 24, 48 and 72 hrs after injection. Whole-body images were obtained with storage phosphor screen. The organs, blood, abscesses muscles were removed, weighed and counted with a γ counter. The percentage of uptake by organs and per gram tissues and abscess/normal tissue (%ID/g) concentration ratios were calculated. The abscesses in mice were well visualized from 24 hrs. The target-to-non-target (T/NT) ratios were 6.71 ± 1.09 (24 hrs), 8.57 ± 0.81 (48 hrs) and 11.41 ± 0.37 (72 hrs) for 131I-labelled anti-MIF McAb group; while in control group of 131I-IgG, T/NT ratios were 4.65 ± 0.63 (24 hrs), 6.44 ± 0.60 (48 hrs) and 8.23 ± 0.35 (72 hrs) (P < 0.05). MIF mRNA expression was threefold increased in inflammatory tissues at 24 hrs compared with normal tissues, and twofold increased at 48 hrs. MIF protein expression was stronger in the inflammatory tissues at 48 hrs after focal inflammation occurred. Our findings suggest that the 131I-labelled anti-MIF McAb appears to be more specific and suitable than 131I-labelled IgG for targeting focal inflammation, which means MIF can be used as a better marker of inflammatory detection.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Umemiya R, Hatta T, Liao M, Tanaka M, Zhou J, Inoue N, Fujisaki K. Haemaphysalis longicornis: Molecular characterization of a homologue of the macrophage migration inhibitory factor from the partially fed ticks. Exp Parasitol 2007; 115:135-42. [PMID: 16987517 DOI: 10.1016/j.exppara.2006.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 07/10/2006] [Accepted: 07/17/2006] [Indexed: 11/16/2022]
Abstract
The macrophage migration inhibitory factor (MIF) has been identified from some vertebrates and invertebrates. MIF is related to inflammation, tumor growth, and angiogenesis in vertebrates. Here, we report the molecular characterization of a homologue of MIF from partially fed Haemaphysalis longicornis. The sequence analysis of the H. longicornis MIF (HlMIF) indicated that its deduced amino acid sequence has an identity of 77% with the MIF of the tick Amblyomma americanum. Western blot analysis using the anti-His-HlMIF antibody showed that HlMIF was up-regulated during blood feeding. Immunohistochemistry showed that the endogenous HlMIF in partially fed ticks was localized to the midgut and epidermal cells. Moreover, the functional assay revealed that the GST-HlMIF inhibited the migration of human monocytes. In conclusion, we consider that HlMIF may facilitate blood feeding by inhibiting host macrophage migration to the feeding lesion or may participate in the proliferation and differentiation of cells in the tick body.
Collapse
Affiliation(s)
- Rika Umemiya
- Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University, Yanagito, Gifu 501-1193, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Zhang F, Di Y, Li J, Shi Y, Zhang L, Wang C, He X, Liu Y, Wan D, Huo K, Gu J. Molecular cloning and characterization of human Aph2 gene, involved in AP-1 regulation by interaction with JAB1. ACTA ACUST UNITED AC 2006; 1759:514-25. [PMID: 17123647 DOI: 10.1016/j.bbaexp.2006.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2006] [Revised: 10/13/2006] [Accepted: 10/13/2006] [Indexed: 12/19/2022]
Abstract
A human Aph2 gene (hAph2) was identified and cloned from a human placenta cDNA library. Bioinformatics analysis revealed hAPH2 protein shares 96% identity with mouse APH2 and contains a zf-DHHC domain (148-210aa), which is always involved in protein-protein or protein-DNA interaction. Differential expression patterns of hAph2 mRNA were observed in normal human tissues. Yeast two-hybrid screening found another hAPH2-interacting protein JAB1. The zf-DHHC domain of hAPH2 and the C-terminal of JAB1 were confirmed to be critical for the interaction. Fused with GFP and expressed in COS-7, NIH/3T3 and SMMC-7721 cell lines, hAPH2 showed predominant distribution in the cytoplasm and co-localized with JAB1 around the nucleus. Furthermore, overexpression of hAPH2 could increase apoptosis of COS-7 cells and negatively regulate JAB1-induced activation of AP-1 in a concentration dependent manner. The expression level of c-jun was also down-regulated by overexpression of hAPH2 in COS-7 cells. These data showed some basic characterization and function of hAph2 (hAPH2), dependent or independent with JAB1.
Collapse
Affiliation(s)
- Fengrui Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute and Cancer Institute of Shanghai Jiaotong University, Shanghai 200032, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ishiguro Y, Ohkawara T, Sakuraba H, Yamagata K, Hiraga H, Yamaguchi S, Fukuda S, Munakata A, Nakane A, Nishihira J. Macrophage migration inhibitory factor has a proinflammatory activity via the p38 pathway in glucocorticoid-resistant ulcerative colitis. Clin Immunol 2006; 120:335-41. [PMID: 16807115 DOI: 10.1016/j.clim.2006.05.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Revised: 05/13/2006] [Accepted: 05/16/2006] [Indexed: 01/05/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that has potent anti-steroid effects and might be implicated in the pathogenesis of Ulecrative colitis (UC). We defined the functional role of MIF in the glucocorticoid (GC)-resistant inflammatory response in UC. Twenty-four colonic samples were obtained from GC responsive cases, GC refractory cases, Crohn's disease and controls. LPMC were isolated from surgical specimens. MIF was strongly expressed at mRNA levels in refractory cases rather than responsive cases with UC and controls. IL-8 production from LPMC was significantly reduced by GC addition in responsive cases but not in refractory cases. In refractory cases, anti-MIF Ab ameliorated GC-resistant IL-8 production and p38-MAPK activity of LPMC. In addition, p38-MAPK antagonist SB230580 also ameliorated GC-resistant IL-8 production. These results suggest that MIF has an additional proinflammatory activity through the p38-MAPK pathway in GC-resistant UC.
Collapse
Affiliation(s)
- Yoh Ishiguro
- First Department of Internal Medicine, Hirosaki University School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Du J, Yu Y, Tu H, Chen H, Xie X, Mou C, Feng K, Zhang S, Xu A. New insights on macrophage migration inhibitory factor: Based on molecular and functional analysis of its homologue of Chinese amphioxus. Mol Immunol 2006; 43:2083-8. [PMID: 16563509 DOI: 10.1016/j.molimm.2005.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2005] [Revised: 12/13/2005] [Accepted: 12/14/2005] [Indexed: 11/24/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an intricate cytokine. Many questions about it are not fully resolved. In order to identify the role of MIF in Chinese amphioxus, its genomic organization, transcription pattern and enzymatic activity were studied. It's found that MIF has multi-copy gene number in the Chinese amphioxus genome and special transcription pattern in reproductive organs. Interestingly, the recombinant Bbt-MIF has tantomerase and redox activity, but fails to utilize GSH to reduce insulin instead of DTT, strikingly different from MIF in mammalian. All these results indicate that MIF gene must have undergone important changes in structure and function during the transition of invertebrate/vertebrate and might exert important role in this primitive species, which may be quite different from those found in vertebrate.
Collapse
Affiliation(s)
- Jingchun Du
- State Key Laboratory of Biocontrol, Department of Biochemistry, Open Laboratory for Marine Functional Genomics of State High-Tech Development Program, Sun Yat-Sen University, Guangzhou 510275, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bernhagen J. Macrophage migration and function: from recruitment in vascular disease to redox regulation in the immune and neuroendocrine networks. Antioxid Redox Signal 2005; 7:1182-8. [PMID: 16115021 DOI: 10.1089/ars.2005.7.1182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Burger-Kentischer A, Finkelmeier D, Thiele M, Schmucker J, Geiger G, Tovar GEM, Bernhagen J. Binding of JAB1/CSN5 to MIF is mediated by the MPN domain but is independent of the JAMM motif. FEBS Lett 2005; 579:1693-701. [PMID: 15757663 DOI: 10.1016/j.febslet.2005.01.080] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 01/07/2005] [Accepted: 01/31/2005] [Indexed: 11/26/2022]
Abstract
Macrophage migration inhibitory factor (MIF) binds to c-Jun activation domain binding protein-1 (JAB1)/subunit 5 of COP9 signalosome (CSN5) and modulates cell signaling and the cell cycle through JAB1. The binding domain of JAB1 responsible for binding to MIF is unknown. We hypothesized that the conserved Mpr1p Pad1p N-terminal (MPN) domain of JAB1 may mediate binding to MIF. In fact, yeast two hybrid (YTH) and in vitro translation/coimmunoprecipitation (CoIP) analysis showed that a core MPN domain, which did not cover the functional JAB1/MPN/Mov34 metalloenzyme (JAMM) deneddylase sequence, binds to MIF comparable to full-length JAB1. YTH and pull-down analysis in conjunction with nanobead affinity matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry demonstrated that MIF(50-65) and MPN are sufficient to mediate MIF-JAB1 interaction, respectively. Finally, endogenous CoIP of MIF-CSN6 complexes from mammalian cells demonstrated that MPN is responsible for MIF-JAB1 binding in vivo, and, as CSN6 does not contain a functional JAMM motif, confirmed that the interaction does not require JAMM.
Collapse
Affiliation(s)
- Anke Burger-Kentischer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Nobelstrasse 12, D-70569 Stuttgart, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Li Y, Lu C, Xing G, Zhu Y, He F. Macrophage migration inhibitory factor directly interacts with hepatopoietin and regulates the proliferation of hepatoma cell. Exp Cell Res 2004; 300:379-87. [PMID: 15475002 DOI: 10.1016/j.yexcr.2004.07.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 07/19/2004] [Indexed: 01/01/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a pluripotent cytokine involved in inflammation and immune responses as well as in growth factor-dependent cell proliferation, cell cycle, angiogenesis, and tumorigenesis. Several studies have documented MIF expression in the sera following hepatic resection or in the course of liver cancer progression, but there is a paucity of information regarding the effect of MIF on hepatoma cells and relating mechanisms. In this paper, by [3H] thymidine incorporation, we found that exogenously added MIF could promote the proliferation of HepG2 in a dose-dependent manner. Hepatopoietin (HPO), as a liver-specific regeneration augmenter, could be induced by the expression of MIF in hepatoma cells. The activity of HPO promoter was increased, and its levels were enhanced after MIF was overexpressed in hepatoma cells. The similarities between HPO and MIF in structure and action led us to investigate their interaction and the inducing biological significance. Using yeast two-hybrid identification, we found that HPO interacted with MIF in yeast cells, and their binding ability was higher than that between HPO and JAB1 (Jun activation domain binding protein) or MIF and JAB1 in yeast cells. Their interaction was further verified by His pull-down assay in vitro and coimmunoprecipitation experiment in vivo. They were colocalized in the cytoplasm. Both HPO and MIF could bind to JAB1 and modulate the AP-1 pathway. When HPO and MIF were cotransfected into HepG2 cells, the binding activity of MIF to JAB1 was reduced, and the activity of AP-1 was improved. In contrast, MIF overexpressed in HepG2 was unable to interfere with the binding activity of HPO to JAB1, but its potentiation on AP-1 activity was reduced significantly. Taken together, these results indicate that MIF plays an important role in the proliferation of hepatoma cells, and the effect of MIF is in concert with HPO.
Collapse
Affiliation(s)
- Yingxian Li
- Laboratory of Systems Biology, Beijing Institute of Radiation Medicine, Chinese Human Genome Center at Beijing, Beijing 100850, China
| | | | | | | | | |
Collapse
|
40
|
Sasaki S, Nishihira J, Ishibashi T, Yamasaki Y, Obikane K, Echigoya M, Sado Y, Ninomiya Y, Kobayashi K. Transgene of MIF induces podocyte injury and progressive mesangial sclerosis in the mouse kidney. Kidney Int 2004; 65:469-81. [PMID: 14717917 DOI: 10.1111/j.1523-1755.2004.00394.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recent evidence suggests that macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that plays a pathogenic role in glomerulonephritis. Renal expression of MIF is up-regulated in infiltrating and intrinsic renal cells, which include glomerular epithelial cells. The aim of the current study was to further clarify the role of MIF produced by podocytes in the process of renal disease. METHODS We generated transgenic mice carrying a murine MIF cDNA driven by cytomegalovirus enhancer and beta-actin/beta-globin promoter, a hybrid promoter transactivated in podocytes in vivo. RESULTS MIF expression was markedly up-regulated in podocytes in neonatal and adult transgenic kidneys. A longitudinal study of the MIF transgenic mice demonstrated a progressive matrix increase in mesangium accompanied by collagen IV accumulation, representing no significant glomerular cell hypercellularity. The glomeruli in transgenic kidney were not accompanied by influx of macrophages and T cells at the early stage of disease progression. Although a significant number of the mice showing higher expression of MIF died from renal failure at 8 weeks, most of them survived with significant proteinuria and progressive renal failure. Podocytes of transgenic mice frequently underwent characteristic ultrastructural changes, such as cell flattening, contracted foot processes, and villous transformation. In addition, immunohistochemical expression of synaptopodin, an actin-associated protein distributed in differentiated podocyte foot process, was significantly attenuated in transgenic kidney. CONCLUSION Our results indicate that podocyte-expressed MIF could induce an injury of podocytes themselves, thereby accelerating the progression of glomerulosclerosis and leading to end-stage renal failure.
Collapse
Affiliation(s)
- Satoshi Sasaki
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Garai J, Adlercreutz H. Estrogen-inducible uterine flavonoid binding sites: is it time to reconsider? J Steroid Biochem Mol Biol 2004; 88:377-81. [PMID: 15145447 DOI: 10.1016/j.jsbmb.2004.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2003] [Accepted: 01/13/2004] [Indexed: 11/21/2022]
Abstract
Epidemiological data support the beneficial effect of plant flavonoids on human health including anti-inflammatory and cancer preventing actions. The phytoestrogen flavonoids might interfere with estrogen action. The possible relations between the steroid- and the flavonoid-signalling in animal and plant cells have been addressed in numerous studies in the past decade. In search for possible sites of conjunction between these phenomena the post-receptor targets must not be disregarded. The estrogen-inducible type II estrogen binding sites of rat uteri have first been reported 25 years ago by Clark and coworkers [Biochem. Biophys. Res. Commun. 81 (1978) 1]. These sites are known to bind catecholic flavonoids with considerable affinity. Behaviour of the tyrosinase-like enzymatic activity associated with these sites appeared reminiscent to the recently described dopachrome oxidase or tautomerase activity exhibited by the cytokine macrophage migration inhibitory factor (MIF) inasmuch as it also accepts a broad range of catecholic melanogenic precursors. Therefore we assessed, whether the known type II ligand flavonoids interfere with the MIF tautomerase. We report here, that luteolin and quercetin have a biphasic effect on the enol-keto conversion of phenylpyruvate mediated by MIF tautomerase. We also demonstrate the presence of MIF immunoreactivity by Western blotting in rat uterine nuclear extracts prepared according to the method that yields high type II binding activity. These data support the possible participation of MIF in type II estrogen binding phenomena.
Collapse
Affiliation(s)
- János Garai
- Department of Pathophysiology, Medical School, University of Pécs, Szigeti u. 12., Pécs H-7624, Hungary.
| | | |
Collapse
|
42
|
Abstract
Cytokines are essential effector molecules of innate immunity that initiate and coordinate the cellular and humoral responses aimed, for example, at the eradication of microbial pathogens. Discovered in the late 1960s as a product of activated T cells, the cytokine macrophage migration inhibitory factor (MIF) has been discovered recently to carry out important functions as a mediator of the innate immune system. Constitutively expressed by a broad spectrum of cells and tissues, including monocytes and macrophages, MIF is rapidly released after exposure to microbial products and pro-inflammatory mediators, and in response to stress. After it is released, MIF induces pro-inflammatory biological responses that act as a regulator of immune responses. MIF activates the extracellular signal-regulated kinase 1 (ERK1)/ERK2–mitogen-activated protein kinase pathway, inhibits the activity of JUN activation domain-binding protein 1 (JAB1) — a co-activator of the activator protein 1 (AP1) — upregulates the expression of Toll-like receptor 4 to promote the recognition of endotoxin-expressing bacterial pathogens, sustains pro-inflammatory function by inhibiting p53-dependent apoptosis of macrophages and counter-regulates the immunosuppressive effects of glucocorticoids on immune cells. As a pro-inflammatory mediator, MIF has been shown to be implicated in the pathogenesis of severe sepsis and septic shock, acute respiratory distress syndrome, and several other inflammatory and autoimmune diseases, including rheumatoid arthritis, glomerulonephritis and inflammatory bowel diseases. Given its crucial role as a regulator of innate and acquired immunity, pharmacological or immunological modulation of MIF activity might offer new treatment opportunities for the management of acute and chronic inflammatory diseases.
For more than a quarter of a century, macrophage migration inhibitory factor (MIF) has been a mysterious cytokine. In recent years, MIF has assumed an important role as a pivotal regulator of innate immunity. MIF is an integral component of the host antimicrobial alarm system and stress response that promotes the pro-inflammatory functions of immune cells. A rapidly increasing amount of literature indicates that MIF is implicated in the pathogenesis of sepsis, and inflammatory and autoimmune diseases, suggesting that MIF-directed therapies might offer new treatment opportunities for human diseases in the future.
Collapse
Affiliation(s)
- Thierry Calandra
- Division of Infectious Diseases, Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland.
| | | |
Collapse
|
43
|
Nguyen MT, Beck J, Lue H, Fünfzig H, Kleemann R, Koolwijk P, Kapurniotu A, Bernhagen J. A 16-residue peptide fragment of macrophage migration inhibitory factor, MIF-(50-65), exhibits redox activity and has MIF-like biological functions. J Biol Chem 2003; 278:33654-71. [PMID: 12796500 DOI: 10.1074/jbc.m301735200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that participates in the host inflammatory response. A Cys-Xaa-Xaa-Cys (CXXC)-based thiol-protein oxidoreductase activity of MIF is associated with certain biological functions. Peptides spanning the CXXC region of thiol-protein oxidoreductases retain some biochemical properties of the full-length protein. We report on the characterization of CXXC-spanning MIF-(50-65) and its serine variant, C57S/C60S-MIF-(50-65). Following disulfide-mediated cyclization, MIF-(50-65) adapted a beta-turn conformation comparable with that of beta-turn-containing cyclo-57,60-[Asp57,Dap60]MIF-(50-65). MIF-(50-65) had a redox potential E'0 of -0.258 V and formed mixed disulfides with glutathione and cysteine. MIF-(50-65) but not C57S/C60S-MIF-(50-65) had oxidoreductase activity in vitro. Intriguingly, MIF-(50-65) exhibited MIF-like cellular activities. The peptide but not its variant had glucocorticoid overriding and proliferation-enhancing activity and stimulated ERK1/2 phosphorylation. MIF-(50-65) and its variant bound to the MIF-binding protein JAB1 and enhanced cellular levels of p27Kip1. As the peptide and its variant were endocytosed at similar efficiency, sequence 50-65 appears sufficient for the JAB1-related effects of MIF, whereas other activities require CXXC. Cyclo-57,60-[Asp57,Dap60]MIF-(50-65) activated ERK1/2, indicating that CXXC-dependent disulfide and beta-turn formation is associated with an activity-inducing conformation. We conclude that CXXC and sequence 50-65 are critical for the activities of MIF. MIF-(50-65) is a surprisingly short sequence with MIF-like functions that could be an excellent molecular template for MIF therapeutics.
Collapse
Affiliation(s)
- Mai Tuyet Nguyen
- Division of Biochemistry and Molecular Cell Biology, Institute of Biochemistry, University Hospital RWTH Aachen, D-52074 Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Iwaki T, Sugimura M, Nishihira J, Matsuura T, Kobayashi T, Kanayama N. Recombinant adenovirus vector bearing antisense macrophage migration inhibitory factor cDNA prevents acute lipopolysaccharide-induced liver failure in mice. J Transl Med 2003; 83:561-70. [PMID: 12695559 DOI: 10.1097/01.lab.0000062857.26210.ef] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine involved in delayed hypersensitivity and cellular immunity. MIF also acts as a proinflammatory cytokine and counterregulates the anti-inflammatory effects of glucocorticoids. Exogenous gene transfer mediated by adenovirus is useful to study a particular molecular function as well as to develop gene therapy strategies. A recombinant adenovirus containing sense and antisense murine MIF (mMIF) cDNA inserts was constructed using a cosmid-terminal protein complex method. The sense mMIF adenovirus (AxCA-mMIFS) efficiently induced mMIF in COS-7 cells that endogenously lack mMIF in a dose-dependent manner. In contrast, the antisense mMIF adenovirus (AxCA-mMIFAS) inhibited the expression of mMIF in NIH3T3 cells in a dose-dependent manner. To assess the pathophysiologic role of MIF in acute liver failure, we induced acute onset of liver damage in mice (male Jcl:ICR) by a combined treatment of Bacille Calmette-Guerin (BCG) and lipopolysaccharide (LPS). mMIF level in the liver of mice infected with AxCA-mMIFAS showed a significant reduction in MIF production in response to BCG-LPS compared with mice treated without viral infection and with AxCA-mMIFS. In addition, the immunohistochemical staining demonstrated that F4/80 antigen on macrophage was enhanced in liver infected with AxCA-mMIFS but reduced in liver infected with AxCA-mMIFAS. The staining intensity is correlated with the mMIF antigen level in liver tissue. The survival rate of mice infected with AxCA-mMIFAS was significantly higher than that of mice treated with PBS and infected with AxCA-LacZ in BCG-LPS. These results suggest that inhibition of MIF production, using recombinant adenovirus bearing the antisense MIF gene, reduced the mortality rate in BCG-LPS-induced liver failure in mice. This finding might aid in the further development of gene therapy targeting MIF.
Collapse
Affiliation(s)
- Takayuki Iwaki
- Department of Obstetrics and Gynecology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Morand EF, Bucala R, Leech M. Macrophage migration inhibitory factor: an emerging therapeutic target in rheumatoid arthritis. ARTHRITIS AND RHEUMATISM 2003; 48:291-9. [PMID: 12571836 DOI: 10.1002/art.10728] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Eric F Morand
- Monash Medical Centre, Clayton, Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
46
|
Lefranc F, Chevalier C, Vinchon M, Dhellemmes P, Schüring MP, Kaltner H, Brotchi J, Ruchoux MM, Gabius HJ, Salmon I, Kiss R. Characterization of the levels of expression of retinoic acid receptors, galectin-3, macrophage migration inhibiting factor, and p53 in 51 adamantinomatous craniopharyngiomas. J Neurosurg 2003; 98:145-53. [PMID: 12546363 DOI: 10.3171/jns.2003.98.1.0145] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Craniopharyngiomas are histopathologically defined as benign tumors that can behave very aggressively at the clinical level. They can originate from different types of embryonal epithelial tissue in which correct spatiotemporal regulation has been disrupted at the effector production level. The goal of this study was to determine the efficacy of using selected biological markers to distinguish between recurring and nonrecurring craniopharyngiomas. METHODS The authors used computer-assisted microscopy to determine quantitatively the immunohistochemical levels of expression of selected markers, including retinoic acid receptors (RARs), as response elements to retinoic acid in a series of 51 adamantinomatous craniopharyngiomas. These tumors may also originate as the result of physiological defects in the apoptosis-mediated elimination of embryological remnants of epithelial tissue. Galectin-3, p53, and the macrophage migration inhibiting factor (MIF) are known to play crucial roles in these processes. The authors quantitatively determined the levels of expression of these substances in this series of 51 craniopharyngiomas. The data show that all craniopharyngiomas were immunoreactive for RARalpha, whereas their immunoreactivity for RARbeta and RARgamma varied dramatically from one case to another. Craniopharyngiomas with low levels of RARbeta and high levels of RARgamma are more likely to recur than those with higher levels of RARbeta and lower levels of RARgamma. Rapidly recurring craniopharyngiomas also show significantly lower levels of expression of galectin-3 and MIF than nonrecurring or slowly recurring cases. Few tumors exhibited p53 immunopositivity. CONCLUSIONS The data indicate that even in the so-called adamantinomatous group of craniopharyngiomas, several subgroups with different clinical behavior patterns can be identified on the basis of differentiation markers relating mainly to the presence or absence of RARbeta and RARgamma.
Collapse
Affiliation(s)
- Florence Lefranc
- Department of Neurosurgery, Erasmus University Hospital and Laboratory of Histopathology, Faculty of Medicine, Free University of Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zang X, Taylor P, Wang JM, Meyer DJ, Scott AL, Walkinshaw MD, Maizels RM. Homologues of human macrophage migration inhibitory factor from a parasitic nematode. Gene cloning, protein activity, and crystal structure. J Biol Chem 2002; 277:44261-7. [PMID: 12221083 DOI: 10.1074/jbc.m204655200] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cytokines are the molecular messengers of the vertebrate immune system, coordinating the local and systemic immune responses to infective organisms. We report here functional and structural data on cytokine-like proteins from a eukaryotic pathogen. Two homologues of the human cytokine macrophage migration inhibitory factor (MIF) have been isolated from the parasitic nematode Brugia malayi. Both molecules (Bm-MIF-1 and Bm-MIF-2) show parallel functions to human MIF. They are chemotactic for human monocytes and activate them to produce IL-8, TNF-alpha, and endogenous MIF. The human and nematode MIF homologues share a tautomerase enzyme activity, which is in each case abolished by the mutation of the N-terminal proline residue. The crystal structure of Bm-MIF-2 at 1.8-A resolution has been determined, revealing a trimeric assembly with an inner pore created by beta-stranded sheets from each subunit. Both biological activity and crystal structure reveal remarkable conservation between a human cytokine and its parasite counterpart despite the considerable phylogenetic divide among these organisms. The strength of the similarity implies that MIF-mediated pathways play an important role in nematode immune evasion strategies.
Collapse
Affiliation(s)
- Xingxing Zang
- Institute of Cell, Animal & Population Biology, University of Edinburgh, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
48
|
Hathout Y, Riordan K, Gehrmann M, Fenselau C. Differential protein expression in the cytosol fraction of an MCF-7 breast cancer cell line selected for resistance toward melphalan. J Proteome Res 2002; 1:435-42. [PMID: 12645915 DOI: 10.1021/pr020006i] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Analysis of differential protein expression in the cytosol of melphalan-resistant and -susceptible MCF-7 cell lines has been carried out using a combination of two-dimensional gel electrophoresis, mass spectrometry, and bioinformatics. Comparison of multiple digitized gel arrays detected several spots as candidates for differentially expressed proteins in melphalan-resistant MCF-7 cells. The up-regulated proteins included retinoic acid binding protein II, an isoform of the macrophage migration inhibition factor, and other unidentified proteins. The down-regulated proteins included calreticulin, cyclophin A, and an isoform of the 27 kD heat shock protein. Correlation of the differential expression of some of the proteins with acquired resistance of MCF7 cells to melphalan is discussed.
Collapse
Affiliation(s)
- Yetrib Hathout
- Department of Chemistry and Biochemistry, University of Maryland, College Park Maryland 20742, USA.
| | | | | | | |
Collapse
|
49
|
Lue H, Kleemann R, Calandra T, Roger T, Bernhagen J. Macrophage migration inhibitory factor (MIF): mechanisms of action and role in disease. Microbes Infect 2002; 4:449-60. [PMID: 11932196 DOI: 10.1016/s1286-4579(02)01560-5] [Citation(s) in RCA: 272] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a unique cytokine and critical mediator of host defenses with a role in septic shock and chronic inflammatory and autoimmune diseases. Its mechanism of action is incompletely understood. Here, we attempt to correlate current knowledge on the molecular pathways of MIF activity with its functions in immunity and disease.
Collapse
Affiliation(s)
- Hongqi Lue
- Laboratory of Biochemistry, Institute for Interfacial Engineering, University of Stuttgart, 70569 Stuttgart, Germany
| | | | | | | | | |
Collapse
|
50
|
Chu J, Jeffries S, Norton JE, Capobianco AJ, Bresnick EH. Repression of activator protein-1-mediated transcriptional activation by the Notch-1 intracellular domain. J Biol Chem 2002; 277:7587-97. [PMID: 11739397 DOI: 10.1074/jbc.m111044200] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Developmental decisions that control cell fate are commonly regulated by the Notch signaling pathway. Activation of transmembrane Notch receptors results in proteolytic liberation of the intracellular domain of Notch, which translocates into the nucleus, binds a repressor (C promoter binding factor 1/RBP-Jkappa, Su(H), and Lag-1 (CSL)), and induces target genes. We found that the intracellular domain of human Notch-1 (NIC-1) represses activator protein-1 (AP-1)-mediated transactivation. Because numerous genes that control immune and inflammatory responses are AP-1-dependent and Notch regulates immune cell function, we investigated the underlying molecular mechanisms. Repression of AP-1 by NIC-1 did not represent a general inhibitory effect on transcription because nuclear factor kappaB-dependent transcription and transcription driven by a constitutive promoter and enhancer were not affected by NIC-1. The physiological relevance of the repression was supported by the facts that repression was apparent in multiple cell lines, endogenous AP-1 target genes were repressed, and similar concentrations of NIC-1 were required for CSL-dependent activation and AP-1 repression. The RBP-Jkappa-associated molecule domain of NIC-1 that mediates CSL binding and distinct sequences necessary for transactivation were required for repression. However, there was not a strict correlation between the sequence requirements for CSL-dependent activation and AP-1 repression. Repression correlated with predominant nuclear localization of NIC-1 and was not accompanied by disruption of c-Jun amino-terminal kinase-dependent signaling events required for AP-1 activation or by defective AP-1 DNA binding activity. These results provide evidence for negative cross-talk between Notch and AP-1, which may have important consequences for controlling diverse biological processes.
Collapse
Affiliation(s)
- Jianlin Chu
- Department of Pharmacology, Molecular and Cellular Pharmacology Program, University of Wisconsin Medical School, 387 Medical Sciences Center, 1300 University Ave., Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|