1
|
Essien SA, Ahuja I, Eisenhoffer GT. Apoptotic extracellular vesicles carrying Mif regulate macrophage recruitment and compensatory proliferation in neighboring epithelial stem cells during tissue maintenance. PLoS Biol 2024; 22:e3002194. [PMID: 39495793 DOI: 10.1371/journal.pbio.3002194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/27/2024] [Indexed: 11/06/2024] Open
Abstract
Apoptotic cells can signal to neighboring cells to stimulate proliferation and compensate for cell loss to maintain tissue homeostasis. While apoptotic cell-derived extracellular vesicles (AEVs) can transmit instructional cues to mediate communication with neighboring cells, the molecular mechanisms that induce cell division are not well understood. Here, we show that macrophage migration inhibitory factor (Mif)-containing AEVs regulate compensatory proliferation via ERK signaling in epithelial stem cells of larval zebrafish. Time-lapse imaging showed efferocytosis of AEVs from dying epithelial stem cells by healthy neighboring stem cells. Proteomic and ultrastructure analysis of purified AEVs identified Mif localization on the AEV surface. Pharmacological inhibition or genetic mutation of Mif, or its cognate receptor CD74, decreased levels of phosphorylated ERK and compensatory proliferation in the neighboring epithelial stem cells. Disruption of Mif activity also caused decreased numbers of macrophages patrolling near AEVs, while depletion of the macrophage lineage resulted in a reduced proliferative response by the epithelial stem cells. We propose that AEVs carrying Mif directly stimulate epithelial stem cell repopulation and guide macrophages to cell non-autonomously induce localized proliferation to sustain overall cell numbers during tissue maintenance.
Collapse
Affiliation(s)
- Safia A Essien
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ivanshi Ahuja
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - George T Eisenhoffer
- Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
2
|
Song AT, Sindeaux RHM, Li Y, Affia H, Agnihotri T, Leclerc S, van Vliet PP, Colas M, Guimond JV, Patey N, Feulner L, Joyal JS, Haddad E, Barreiro L, Andelfinger G. Developmental role of macrophages modeled in human pluripotent stem cell-derived intestinal tissue. Cell Rep 2024; 43:113616. [PMID: 38150367 DOI: 10.1016/j.celrep.2023.113616] [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] [Received: 10/25/2023] [Revised: 11/22/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023] Open
Abstract
Macrophages populate the embryo early in gestation, but their role in development is not well defined. In particular, specification and function of macrophages in intestinal development remain little explored. To study this event in the human developmental context, we derived and combined human intestinal organoid and macrophages from pluripotent stem cells. Macrophages migrate into the organoid, proliferate, and occupy the emerging microanatomical niches of epithelial crypts and ganglia. They also acquire a transcriptomic profile similar to that of fetal intestinal macrophages and display tissue macrophage behaviors, such as recruitment to tissue injury. Using this model, we show that macrophages reduce glycolysis in mesenchymal cells and limit tissue growth without affecting tissue architecture, in contrast to the pro-growth effect of enteric neurons. In short, we engineered an intestinal tissue model populated with macrophages, and we suggest that resident macrophages contribute to the regulation of metabolism and growth of the developing intestine.
Collapse
Affiliation(s)
- Andrew T Song
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada.
| | - Renata H M Sindeaux
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Meakins Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Yuanyi Li
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada
| | - Hicham Affia
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada
| | - Tapan Agnihotri
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | | | | | - Mathieu Colas
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada
| | - Jean-Victor Guimond
- CLSC des Faubourgs, CIUSSS du Centre-Sud-de-l'Ile-de-Montréal, Montréal, QC, Canada
| | - Natalie Patey
- Department of Pathology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Lara Feulner
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada
| | - Jean-Sebastien Joyal
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Elie Haddad
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada
| | - Luis Barreiro
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Genetics Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Gregor Andelfinger
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada; Département de Pédiatrie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
3
|
Su Y, Zhang X, Liang Y, Sun J, Lu C, Huang Z. Integrated analysis of single-cell RNA-seq and bulk RNA-seq to unravel the molecular mechanisms underlying the immune microenvironment in the development of intestinal-type gastric cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166849. [PMID: 37591405 DOI: 10.1016/j.bbadis.2023.166849] [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] [Received: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
Intestinal-type gastric cancer (IGC) is the most frequent type of gastric cancer in high-incidence populations. The early stages of IGC growth successively include nonatrophic gastritis (NAG), chronic atrophic gastritis (CAG) and intestinal metaplasia (IM). However, the mechanisms of IGC development through these stages remain unclear. For this study, single-cell RNA-seq data related to IGC were downloaded from the GEO database, and immune cells of the tumor microenvironment (TME) were annotated using R software. Changes in the proportion of immune cells and altered cell-to-cell interactions were explored at different disease stages using R software, with a focus on plasma cells. Additionally, IGC samples from the TCGA database were used for immune cell infiltration analysis, and a Cox proportional risk regression model was constructed to identify possible prognostic genes. The results indicated that for precancerous lesions, interactions between immune cells were mainly dominated by chemokines to stimulate the infiltration and activation of immune cells. In tumors, intercellular movement of upregulated molecules and amplified signals were associated with the tumor necrosis factor family and immunosuppression to escape immune surveillance and promote tumor growth. Regarding prognostic analysis, IGLC3, IGLV1-44, IGKV1-16, IGHV3-21, IGLV1-51, and IGLV3-19 were found to be novel biomarkers for IGC. Our analysis of the IGC single-cell atlas together with bulk transcriptome data contributes to understanding TME heterogeneity at the molecular level during IGC development and provides insights for elucidating the mechanism of IGC and discovering novel targets for precise therapy.
Collapse
Affiliation(s)
- Yongjian Su
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xiaoqing Zhang
- School of Basic Medicine, Guangdong Medical University, Dongguan, China
| | - Youcheng Liang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jianbo Sun
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Chengyu Lu
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
4
|
Spiller L, Manjula R, Leissing F, Basquin J, Bourilhon P, Sinitski D, Brandhofer M, Levecque S, Gerra S, Sabelleck B, Zhang L, Feederle R, Flatley A, Hoffmann A, Panstruga R, Bernhagen J, Lolis E. Plant MDL proteins synergize with the cytokine MIF at CXCR2 and CXCR4 receptors in human cells. Sci Signal 2023; 16:eadg2621. [PMID: 37988455 DOI: 10.1126/scisignal.adg2621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 10/27/2023] [Indexed: 11/23/2023]
Abstract
Mammalian macrophage migration inhibitory factor (MIF) and its paralog, D-dopachrome tautomerase, are multifunctional inflammatory cytokines. Plants have orthologous MIF and D-dopachrome tautomerase-like (MDL) proteins that mimic some of the effects of MIF on immune cells in vitro. We explored the structural and functional similarities between the three Arabidopsis thaliana MDLs and MIF. X-ray crystallography of the MDLs revealed high structural similarity between MDL and MIF homotrimers and suggested a potential explanation for the lack of tautomerase activity in the MDLs. MDL1 and MDL2 interacted with each other and with MIF in vitro, in yeast, and in plant leaves and formed hetero-oligomeric complexes with MIF in vitro. The MDLs stimulated signaling through the MIF receptors CXCR2 or CXCR4 and enhanced the responses to MIF in a yeast reporter system, in human neutrophils, and in human lung epithelial cells. Pharmacological inhibitors that disrupted MIF activity or prevented the formation of MIF-MDL hetero-oligomers blocked the observed synergism. These findings demonstrate that MDLs can enhance cellular responses to MIF, which may have functional implications in tissues exposed to MDLs from the diet or environment.
Collapse
Affiliation(s)
- Lukas Spiller
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Ramu Manjula
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Franz Leissing
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Jerome Basquin
- Department of Structural Cell Biology and Crystallization Facility, Max-Planck-Institute for Biochemistry, 82152 Martinsried, Germany
| | - Priscila Bourilhon
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Dzmitry Sinitski
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Sophie Levecque
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Simona Gerra
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
| | - Björn Sabelleck
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Lin Zhang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Department of Anesthesiology, LMU University Hospital, 81377 Munich, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Andrew Flatley
- Monoclonal Antibody Core Facility, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Department of Anesthesiology, LMU University Hospital, 81377 Munich, Germany
| | - Ralph Panstruga
- Unit of Plant Molecular Cell Biology, Institute for Biology I, RWTH Aachen University, 52056 Aachen, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität (LMU) München, LMU University Hospital, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Elias Lolis
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
5
|
Xie Y, Xu D, Yan S, Hu X, Chen S, Guo K, Wang J, Chen Q, Guan W. The impact of MIF deficiency on alterations of fecal microbiota in C57BL/6 mice induced by Trichinella spiralis infection. FASEB J 2023; 37:e23202. [PMID: 37732633 DOI: 10.1096/fj.202300179rr] [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] [Received: 02/01/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Trichinellosis caused by Trichinella spiralis (T. spiralis) is a major food-borne parasitic zoonosis worldwide. Prevention of trichinellosis is an effective strategy to improve patient quality of life. Macrophage migration inhibitory factor (MIF) is closely related to the occurrence and development of several parasitic diseases. Studying the impact of MIF deficiency (Mif-/- ) on the alterations in host fecal microbiota due to T. spiralis infection may contribute to proposing a novel dual therapeutic approach for trichinellosis. To reveal the diversity and differences in fecal microbial composition, feces were collected from T. spiralis-uninfected and T. spiralis-infected wild-type (WT) and MIF knockout (KO) C57BL/6 mice at 0, 7, 14, and 35 days post-infection (dpi), and the samples were sent for 16S rRNA amplicon sequencing on the Illumina NovaSeq platform. Flow cytometry was used to determine the expression levels of IFN-γ and IL-4 in the CD4+ /CD8+ T-cell sets of mouse spleens. The results showed that operational taxonomic unit (OTU) clustering, relative abundance of microbial composition, alpha diversity, and beta diversity exhibited significant changes among the eight groups. The LEfSe analysis selected several potential biomarkers at the genus or species level, including Akkermansia muciniphila, Lactobacillus murinus, Coprococcus catus, Firmicutes bacterium M10_2, Parabacteroides sp. CT06, and Bacteroides between the KTs and WTs groups. The predicted bacterial functions of the fecal microbiota were mainly involved in metabolism, such as the metabolism of carbohydrates, amino acids, energy, cofactors, vitamins, nucleotides, glycans, and lipids. Flow cytometry revealed an increased CD3+ CD8- /CD3+ CD8+ T-cell ratio and increased IFN-γ and IL-4 levels in CD3+ CD8- T-cell sets from WT and MIF KO mice at 7 dpi. The results indicated that both MIF KO and infection time have a significant influence on the CD3+ CD8- IFN-γ+ and CD3+ CD8- IL-4+ response in mice after T. spiralis. In conclusion, this research showed alterations of the fecal microbiota and immune response in both WT and MIF KO mice before and after T. spiralis infection. These results revealed a potential role of MIF in regulating the pathogenesis of trichinellosis related to the intestinal microbiota. Importantly, the selected potential biomarkers combined with MIF will also offer a novel therapeutic approach to treat trichinellosis in the future.
Collapse
Affiliation(s)
- Yiting Xie
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Daoxiu Xu
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Siyi Yan
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Xinyi Hu
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Sirui Chen
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Kun Guo
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Jue Wang
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Qinghai Chen
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Wei Guan
- Department of Human Parasitology, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
6
|
Xie Y, Guan W, Zhao Y, Yan S, Guo K, Chen S, Hu X, Shi H, Li J. Deficiency of migration inhibitory factor influences the gut microbiota of C57BL/6 mice infected with Plasmodium berghei ANKA. Front Microbiol 2022; 13:978644. [PMID: 36033889 PMCID: PMC9412183 DOI: 10.3389/fmicb.2022.978644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
Cerebral malaria (CM), as one of the most common complications in severe malaria, has threatened millions of individuals’ neurological health and even their lives. Macrophage migration inhibitory factor (MIF), a pleiotropic proinflammatory factor in humans, seems to be a risk factor for death in patients with CM, but its functional mechanism remains unclear. To verify whether affecting the intestinal microbes of the host was one of the mechanisms by which MIF regulates CM, C57BL/6 mice, including WT + PbA, MIF-KO + PbA and their uninfected controls, were sent for 16S rRNA-based sequencing targeting the V4 region of the intestinal microbiota through the Illumina MiSeq platform. The results showed that OTU clustering, alpha and beta diversity in the four groups involved had evident variation. The relative abundance at different taxonomic levels, especially the dominant intestinal flora, was obviously changed. The LEfSe analysis screened out several biomarkers, including significantly reduced Ligilactobacillus (Lactobacillus murinus) in WPbA mice compared to the WT group and Akkermansia (Akkermansia_muciniphila) in KPbA mice compared to the WPbA group. For MIF KO groups, mice infected with PbA or uninfected showed significant enrichment of producers of short-chain fatty acids, including Dubosiella and Faecalibaculum (Faecalibaculum rodentium) in KPbA, and Lachnospiraceae_NK4A136_group and Firmicutes_bacterium_M10-2 in KO. This study not only further proved the gut microbiota changes in C57BL/6 mice caused by PbA infection, but also found that MIF deletion directly affected the changes in the gut microbiota of C57BL/6 mice before and after PbA infection. This finding reveals a potential mechanism by which MIF regulates CM. Combining MIF with potential microbial biomarkers will provide a promising idea to develop combined drugs for improving CM in the future.
Collapse
|
7
|
Klemke L, De Oliveira T, Witt D, Winkler N, Bohnenberger H, Bucala R, Conradi LC, Schulz-Heddergott R. Hsp90-stabilized MIF supports tumor progression via macrophage recruitment and angiogenesis in colorectal cancer. Cell Death Dis 2021; 12:155. [PMID: 33542244 PMCID: PMC7862487 DOI: 10.1038/s41419-021-03426-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/02/2021] [Accepted: 01/07/2021] [Indexed: 12/19/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream regulator of innate immunity, but its expression is increased in some cancers via stabilization with HSP90-associated chaperones. Here, we show that MIF stabilization is tumor-specific in an acute colitis-associated colorectal cancer (CRC) mouse model, leading to tumor-specific functions and selective therapeutic vulnerabilities. Therefore, we demonstrate that a Mif deletion reduced CRC tumor growth. Further, we define a dual role for MIF in CRC tumor progression. Mif deletion protects mice from inflammation-associated tumor initiation, confirming the action of MIF on host inflammatory pathways; however, macrophage recruitment, neoangiogenesis, and proliferative responses are reduced in Mif-deficient tumors once the tumors are established. Thus, during neoplastic transformation, the function of MIF switches from a proinflammatory cytokine to an angiogenesis promoting factor within our experimental model. Mechanistically, Mif-containing tumor cells regulate angiogenic gene expression via a MIF/CD74/MAPK axis in vitro. Clinical correlation studies of CRC patients show the shortest overall survival for patients with high MIF levels in combination with CD74 expression. Pharmacological inhibition of HSP90 to reduce MIF levels decreased tumor growth in vivo, and selectively reduced the growth of organoids derived from murine and human tumors without affecting organoids derived from healthy epithelial cells. Therefore, novel, clinically relevant Hsp90 inhibitors provide therapeutic selectivity by interfering with tumorigenic MIF in tumor epithelial cells but not in normal cells. Furthermore, Mif-depleted colonic tumor organoids showed growth defects compared to wild-type organoids and were less susceptible toward HSP90 inhibitor treatment. Our data support that tumor-specific stabilization of MIF promotes CRC progression and allows MIF to become a potential and selective therapeutic target in CRC.
Collapse
Affiliation(s)
- Luisa Klemke
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Daria Witt
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Winkler
- Institute of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Richard Bucala
- Departments of Medicine, Pathology, and Epidemiology & Public Health, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | | |
Collapse
|
8
|
The cytokine MIF controls daily rhythms of symbiont nutrition in an animal-bacterial association. Proc Natl Acad Sci U S A 2020; 117:27578-27586. [PMID: 33067391 DOI: 10.1073/pnas.2016864117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The recent recognition that many symbioses exhibit daily rhythms has encouraged research into the partner dialogue that drives these biological oscillations. Here we characterized the pivotal role of the versatile cytokine macrophage migration inhibitory factor (MIF) in regulating a metabolic rhythm in the model light-organ symbiosis between Euprymna scolopes and Vibrio fischeri As the juvenile host matures, it develops complex daily rhythms characterized by profound changes in the association, from gene expression to behavior. One such rhythm is a diurnal shift in symbiont metabolism triggered by the periodic provision of a specific nutrient by the mature host: each night the symbionts catabolize chitin released from hemocytes (phagocytic immune cells) that traffic into the light-organ crypts, where the population of V. fischeri cells resides. Nocturnal migration of these macrophage-like cells, together with identification of an E. scolopes MIF (EsMIF) in the light-organ transcriptome, led us to ask whether EsMIF might be the gatekeeper controlling the periodic movement of the hemocytes. Western blots, ELISAs, and confocal immunocytochemistry showed EsMIF was at highest abundance in the light organ. Its concentration there was lowest at night, when hemocytes entered the crypts. EsMIF inhibited migration of isolated hemocytes, whereas exported bacterial products, including peptidoglycan derivatives and secreted chitin catabolites, induced migration. These results provide evidence that the nocturnal decrease in EsMIF concentration permits the hemocytes to be drawn into the crypts, delivering chitin. This nutritional function for a cytokine offers the basis for the diurnal rhythms underlying a dynamic symbiotic conversation.
Collapse
|
9
|
Harrison LM, Lacher DW, Mammel MK, Leonard SR. Comparative Transcriptomics of Shiga Toxin-Producing and Commensal Escherichia coli and Cytokine Responses in Colonic Epithelial Cell Culture Infections. Front Cell Infect Microbiol 2020; 10:575630. [PMID: 33194815 PMCID: PMC7649339 DOI: 10.3389/fcimb.2020.575630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Ingestion of Shiga toxin-producing Escherichia coli (STEC) can result in a range of illness severity from asymptomatic to hemorrhagic colitis and death; thus risk assessment of STEC strains for human pathogenicity is important in the area of food safety. Illness severity depends in part on the combination of virulence genes carried in the genome, which can vary between strains even of identical serotype. To better understand how core genes are regulated differently among strains and to identify possible novel STEC virulence gene candidates that could be added to the risk assessment repertoire, we used comparative transcriptomics to investigate global gene expression differences between two STEC strains associated with severe illness and a commensal E. coli strain during in vitro intestinal epithelial cell (IEC) infections. Additionally, we compared a wide array of concomitant cytokine levels produced by the IECs. The cytokine expression levels were examined for a pattern representing STEC pathogenicity; however, while one STEC strain appeared to elicit a proinflammatory response, infection by the other strain produced a pattern comparable to the commensal E. coli. This result may be explained by the significant differences in gene content and expression observed between the STEC strains. RNA-Seq analysis revealed considerable disparity in expression of genes in the arginine and tryptophan biosynthesis/import pathways between the STEC strains and the commensal E. coli strain, highlighting the important role some amino acids play in STEC colonization and survival. Contrasting differential expression patterns were observed for genes involved in respiration among the three strains suggesting that metabolic diversity is a strategy utilized to compete with resident microflora for successful colonization. Similar temporal expression results for known and putative virulence genes were observed in the STEC strains, revealing strategies used for survival prior to and after initial adherence to IECs. Additionally, three genes encoding hypothetical proteins located in mobile genetic elements were, after interrogation of a large set of E. coli genomes, determined to likely represent novel STEC virulence factors.
Collapse
Affiliation(s)
- Lisa M Harrison
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - David W Lacher
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Mark K Mammel
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| | - Susan R Leonard
- Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, United States
| |
Collapse
|
10
|
Marcon CF, Ferreira PTM, Franco PS, Ribeiro M, Silva RJ, Sousa RAP, Oliveira CJF, Rodrigues Junior V, Gomes MLM, Lazo Chica JE, Mineo TWP, Mineo JR, Barbosa BF, Ferro EAV, Gomes AO. Macrophage migration inhibitory factor (MIF) and pregnancy may impact the balance of intestinal cytokines and the development of intestinal pathology caused by Toxoplasma gondii infection. Cytokine 2020; 136:155283. [PMID: 32947151 DOI: 10.1016/j.cyto.2020.155283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Toxoplasma gondii (T. gondii) is an intracellular parasite responsible for causing toxoplasmosis. When infection occurs during pregnancy, it can produce severe congenital infection with ocular and neurologic damage to the infant. From the oral infection parasite reaches the intestine, causing inflammatory response, damage in tissue architecture and systemic dissemination. Macrophage migration inhibition factor (MIF) is a cytokine secreted from both immune and non-immune cells, including gut epithelial cells. MIF is described to promote inflammatory responses, to be associated in colitis pathogenesis and also to play role in maintaining the intestinal barrier. The aim of the present study was to evaluate the influence of the pregnancy and MIF deficiency on T. gondii infection in the intestinal microenvironment and to address how these factors can impact on the intestinal architecture and local cytokine profile. For this purpose, small intestine of pregnant and non-pregnant C57BL/6 MIF deficient mice (MIF-/-) and Wild-type (WT) orally infected with 5 cysts of ME-49 strain of T. gondii were collected on day 8th of infection. Intestines were processed for morphological and morphometric analyses, parasite quantification and for cytokines mensuration. Our results showed that the absence of MIF and pregnancy caused an increase in T. gondii infection index. T. gondii immunolocalization demonstrated that segments preferentially infected with T. gondii were duodenum and ileum. The infection caused a reduction in the size of the intestinal villi, whereas, infection associated with pregnancy caused an increase in villi size due to edema caused by the infection. Also, the goblet cell number was increased in the ileum of MIF-/- mice, when compared to the corresponding WT group. Analyses of cytokine production in the small intestine showed that MIF was up regulated in the gut of pregnant WT mice due to infection. Also, infection provoked an intense Th1 response that was more exacerbated in pregnant MIF-/- mice. We also detected that the Th2/Treg response was more pronounced in MIF-/- mice. Altogether, our results demonstrated that pregnancy and MIF deficiency interferes in the balance of the intestinal cytokines and favors a Th1-immflamatory profile, which in turn, impact in the development of pathology caused by T. gondii infection in the intestinal microenvironment.
Collapse
Affiliation(s)
- Camila Ferreira Marcon
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Paula Tatiana Mutão Ferreira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Priscila Silva Franco
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Roberto Augusto Pereira Sousa
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues Junior
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marcos Lucca Moreira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Javier Emílio Lazo Chica
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Angelica Oliveira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Marcon CF, Ferreira PTM, Franco PS, Ribeiro M, Silva RJ, Sousa RAP, Oliveira CJF, Junior VR, Gomes MLM, Chica JEL, Mineo TWP, Mineo JR, Barbosa BF, Ferro EAV, Gomes AO. WITHDRAWN: Macrophage migration inhibitory factor (MIF) and pregnancy may impact the balance of intestinal cytokines and the development of intestinal pathology caused by Toxoplasma gondii infection. Cytokine X 2020; 2:100034. [PMID: 33604559 PMCID: PMC7885889 DOI: 10.1016/j.cytox.2020.100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Camila Ferreira Marcon
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Paula Tatiana Mutão Ferreira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Priscila Silva Franco
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Mayara Ribeiro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Rafaela José Silva
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Roberto Augusto Pereira Sousa
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues Junior
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marcos Lucca Moreira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Javier Emílio Lazo Chica
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - José Roberto Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratório de Imunofisiologia da Reprodução, Instituto de Ciências Biomédicas (ICBIM), Universidade Federal de Uberlândia (UFU), Uberlândia, Brazil
| | - Angelica Oliveira Gomes
- Instituto de Ciências Biológicas e Naturais (ICBN), Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| |
Collapse
|
12
|
Jeffrey MP, MacPherson CW, Mathieu O, Tompkins TA, Green-Johnson JM. Secretome-Mediated Interactions with Intestinal Epithelial Cells: A Role for Secretome Components from Lactobacillus rhamnosus R0011 in the Attenuation of Salmonella enterica Serovar Typhimurium Secretome and TNF-α-Induced Proinflammatory Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:2523-2534. [PMID: 32238458 DOI: 10.4049/jimmunol.1901440] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Recent evidence suggests that lactic acid bacteria communicate with host cells via secretome components to influence immune responses but less is known about gut-pathogen secretomes, impact of lactic acid bacteria secretomes on host-pathogen interactions, and the mechanisms underlying these interactions. Genome-wide microarrays and cytokine profiling were used to interrogate the impact of the Lactobacillus rhamnosus R0011 secretome (LrS) on TNF-α and Salmonella enterica subsp. enterica serovar Typhimurium secretome (STS)-induced outcomes in human intestinal epithelial cells. The LrS attenuated both TNF-α- and STS-induced gene expression involved in NF-κB and MAPK activation, as well as expression of genes involved in other immune-related signaling pathways. Specifically, the LrS induced the expression of dual specificity phosphatase 1 (DUSP1), activating transcription factor 3 (ATF3), and tribbles pseudokinase 3 (TRIB3), negative regulators of innate immune signaling, in HT-29 intestinal epithelial cells challenged with TNF-α or STS. TNF-α- and STS-induced acetylation of H3 and H4 histones was attenuated by the LrS, as was the production of TNF-α- and STS-induced proinflammatory cytokines and chemokines. Interestingly, the LrS induced production of macrophage migration inhibitory factor (MIF), a cytokine involved in host-microbe interactions at the gut interface. We propose that the LrS attenuates proinflammatory mediator expression through increased transcription of negative regulators of innate immune activity and changes in global H3 and H4 histone acetylation. To our knowledge, these findings provide novel insights into the complex multifaceted mechanisms of action behind secretome-mediated interdomain communication at the gut-mucosal interface.
Collapse
Affiliation(s)
- Michael P Jeffrey
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Technical University, Oshawa, Ontario L1G 0C5, Canada; and
| | - Chad W MacPherson
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Olivier Mathieu
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Thomas A Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Julia M Green-Johnson
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Technical University, Oshawa, Ontario L1G 0C5, Canada; and
| |
Collapse
|
13
|
Stošić-Grujičić S, Saksida T, Miljković Đ, Stojanović I. MIF and insulin: Lifetime companions from common genesis to common pathogenesis. Cytokine 2019; 125:154792. [PMID: 31400637 DOI: 10.1016/j.cyto.2019.154792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/01/2019] [Accepted: 07/24/2019] [Indexed: 12/19/2022]
Abstract
Pro-inflammatory nature of macrophage migration inhibitory factor (MIF) has been generally related to the propagation of inflammatory and autoimmune diseases. But this molecule possesses many other peculiar functions, unrelated to the immune system, among which is its supportive role in the post-translational modifications of insulin. In this way MIF enables proper insulin conformation within the pancreatic beta cell and its full activity. The inherent or acquired changes in MIF expression might therefore lead to different insulin processing and initiation of autoimmunity. The relation between MIF and insulin does not stop at this point; these two molecules continue to interact during pathological states characterized by inflammation and insulin resistance. In this context, MIF indirectly and negatively influences insulin action by boosting inflammatory environment and disabling target cells to respond to insulin. On the other side, insulin might interfere with MIF action as well, acting as an anti-inflammatory mediator. Therefore, the proper interaction between MIF and insulin is crucial for maintaining homeostasis, while anti-inflammatory therapies based on the systemic MIF blockage may disturb this balance. This review covers MIF-insulin relationship in the physiological and pathological conditions and discusses the approaches for MIF inhibition and their net effect specifically considering possible impact on insulin misfolding and the possible misinterpretation of previous results due to the discovery of MIF functional homolog D-dopachrome tautomerase.
Collapse
Affiliation(s)
- Stanislava Stošić-Grujičić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
14
|
Macrophage Migration Inhibitory Factor Promotes the Interaction between the Tumor, Macrophages, and T Cells to Regulate the Progression of Chemically Induced Colitis-Associated Colorectal Cancer. Mediators Inflamm 2019; 2019:2056085. [PMID: 31360118 PMCID: PMC6652048 DOI: 10.1155/2019/2056085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022] Open
Abstract
Colitis-associated colorectal cancer (CRC) development has been shown to be related to chronically enhanced inflammation. Macrophage migration inhibitory factor (MIF) is an inflammatory mediator that favors inflammatory cytokine production and has chemotactic properties for the recruitment of macrophages (Møs) and T cells. Here, we investigated the role of MIF in the inflammatory response and recruitment of immune cells in a murine model of chemical carcinogenesis to establish the impact of MIF on CRC genesis and malignancy. We used BALB/c MIF-knockout (MIF-/-) and wild-type (WT) mice to develop CRC by administering intraperitoneal (i.p.) azoxymethane and dextran sodium sulfate in drinking water. Greater tumor burdens were observed in MIF-/- mice than in WT mice. Tumors from MIF-/- mice were histologically identified to be more aggressive than tumors from WT mice. The localization of MIF suggests that it is also involved in cell differentiation. The relative gene expression of il-17, measured by real-time PCR, was higher in MIF-/- CRC mice, compared to the WT CRC and healthy MIF-/- mice. Importantly, compared to the WT intestinal epithelium, lower percentages of tumor-associated Møs were found in the MIF-/- intestinal epithelium. These results suggest that MIF plays a role in controlling the initial development of CRC by attracting Møs to the tumor, which is a condition that favors the initial antitumor responses.
Collapse
|
15
|
Wang C, Zuo B, Wu X. The Role of Macrophage Migration Inhibitory Factor in Remote Ischemic Postconditioning. Can J Cardiol 2019; 35:501-510. [DOI: 10.1016/j.cjca.2018.12.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/08/2018] [Accepted: 12/30/2018] [Indexed: 01/23/2023] Open
|
16
|
Vujicic M, Saksida T, Despotovic S, Bajic SS, Lalić I, Koprivica I, Gajic D, Golic N, Tolinacki M, Stojanovic I. The Role of Macrophage Migration Inhibitory Factor in the Function of Intestinal Barrier. Sci Rep 2018; 8:6337. [PMID: 29679061 PMCID: PMC5910418 DOI: 10.1038/s41598-018-24706-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 04/09/2018] [Indexed: 12/15/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional protein that is involved in the development of gut-related inflammation. To investigate the role of MIF in the function of the intestinal barrier, we have explored intestinal permeability and gut-associated immune response in MIF-deficient (MIF-KO) mice. The absence of MIF provoked impairment of tight and adherens epithelial junctions in the colon through the disturbance of E-cadherin, zonula occludens-1, occludin and claudin-2 expression, which lead to the increase of intestinal barrier permeability. In these circumstances the diversity and content of gut microbiota in MIF-KO mice was considerably different compared to wild type mice. This change in microbiota was accompanied by an increased intestinal IgA concentration and a higher production of pro-inflammatory cytokines TNF and IFN-γ in mesenteric lymph nodes of MIF-KO mice. The forced changes of microbiota executed by antibiotics prevented the "leakage" of the barrier in MIF-KO mice, probably through up-regulation of occludin expression and normalization of cellular pore diameters. In addition, cytokine secretion was normalized after the treatment with antibiotics. These results suggest that MIF participates in the maintenance of physiological microbiota diversity and immunosurveillance, which in turn enables the proper intestinal barrier function.
Collapse
Affiliation(s)
- Milica Vujicic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Department of Immunology, Belgrade, 11060, Serbia
| | - Tamara Saksida
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Department of Immunology, Belgrade, 11060, Serbia
| | - Sanja Despotovic
- Faculty of Medicine, University of Belgrade, Institute of Histology and Embryology, Belgrade, 11000, Serbia
| | - Svetlana Sokovic Bajic
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, 11000, Serbia
| | - Ivana Lalić
- Faculty of Medicine, University of Belgrade, Institute of Histology and Embryology, Belgrade, 11000, Serbia
| | - Ivan Koprivica
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Department of Immunology, Belgrade, 11060, Serbia
| | - Dragica Gajic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Department of Immunology, Belgrade, 11060, Serbia
| | - Natasa Golic
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, 11000, Serbia
| | - Maja Tolinacki
- Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, 11000, Serbia
| | - Ivana Stojanovic
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Department of Immunology, Belgrade, 11060, Serbia.
| |
Collapse
|
17
|
Moudi B, Heidari Z, Mahmoudzadeh-Sagheb H, Hashemi M. Gene polymorphisms of macrophage migration inhibitory factor affect susceptibility to chronic hepatitis B virus infection in an Iranian cohort. Microbiol Immunol 2017; 60:390-6. [PMID: 27079941 DOI: 10.1111/1348-0421.12382] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/04/2016] [Accepted: 04/06/2016] [Indexed: 12/22/2022]
Abstract
Macrophage migration inhibitory factor (MIF), a key proinflammatory mediator, plays important roles in chronic diseases. In this study, an attempt was made to clarify the associations between some functional polymorphisms such as MIF-173 G/C, MIF 95 bp and 189 bp insertion/deletion (I/D) polymorphisms and chronic hepatitis B virus (HBV) infection. Polymorphisms were assessed in 221 HBV patients and 200 normal subjects. MIF-173 G/C and MIF 95 bp and 189 bp I/D polymorphisms were genotyped using PCR-RFLP and PCR, respectively. When allele and genotype frequencies of the variants were compared between patients and controls by the χ(2) test, it was found that the frequency of MIF-173 G/C genotypes differed significantly between patients with chronic HBV and healthy controls (P < 0.05). Carriers of the MIF -173-C allele were at significantly higher risk of HBV infection than carriers of the MIF -173-G allele (P = 0.009, OR = 1.549, 95% CI = 1.114 - 2.155). Moreover, 95 bp I/D polymorphism was not associated with CP and the 185 bp I/D variant was not polymorphic in our group of subjects. The frequency of haplotypes did not differ significantly between groups (χ(2) = 11.391, P = 0.181). Our results suggest that MIF -173 G/C variant increases the risk of HBV in Iranian subjects. Further studies with larger sample sizes and different ethnicities are required to validate our findings.
Collapse
Affiliation(s)
- Bita Moudi
- Infectious Diseases and Tropical Medicine Research Center.,Department of Histology, School of Medicine
| | - Zahra Heidari
- Infectious Diseases and Tropical Medicine Research Center.,Department of Histology, School of Medicine
| | | | - Mohammad Hashemi
- Cellular and Molecular Research Center.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
18
|
No YR, Lee SJ, Kumar A, Yun CC. HIF1α-Induced by Lysophosphatidic Acid Is Stabilized via Interaction with MIF and CSN5. PLoS One 2015; 10:e0137513. [PMID: 26352431 PMCID: PMC4564097 DOI: 10.1371/journal.pone.0137513] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/18/2015] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that has broad effects on immune system and inflammatory response. A growing body of evidence implicates the role of MIF in tumor growth and metastasis. Lysophosphatidic acid (LPA), a bioactive lipid mediator, regulates colon cancer cell proliferation, invasion, and survival through LPA2 receptor. Loss of LPA2 results in decreased expression of MIF in a rodent model of colon cancer, but the mechanism of MIF regulation by LPA is yet to be determined. In this study, we show that LPA transcriptionally regulates MIF expression in colon cancer cells. MIF knockdown decreased LPA-mediated proliferation of HCT116 human adenocarcinoma cells without altering the basal proliferation rates. Conversely, extracellular recombinant MIF stimulated cell proliferation, suggesting that the effect of MIF may in part be mediated through activation of surface receptor. We have shown recently that LPA increases hypoxia-inducible factor 1α (HIF1α) expression. We found that MIF regulation by LPA was ablated by knockdown of HIF1α, indicating that MIF is a transcriptional target of HIF1α. Conversely, knockdown of MIF ablated an increase in HIF1α expression in LPA-treated cells, suggesting a reciprocal relationship between HIF1α and MIF. LPA stimulated co-immunoprecipitation of HIF1α and MIF, indicating that their association is necessary for stabilization of HIF1α. It has been shown previously that CSN9 signalosome subunit 5 (CSN5) interacts with HIF1α to stabilize HIF1α under aerobic conditions. We found that LPA did not alter expression of CSN5, but stimulated its interaction with HIF1α and MIF. Depletion of CSN5 mitigated the association between HIF1α and MIF, indicating that CSN5 acts as a physical link. We suggest that HIF1α, MIF, and CSN5 form a ternary complex whose formation is necessary to prevent degradation of HIF1α under aerobic conditions.
Collapse
Affiliation(s)
- Yi Ran No
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Sei-Jung Lee
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ajay Kumar
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America; Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
19
|
Gordon-Weeks AN, Lim SY, Yuzhalin AE, Jones K, Muschel R. Macrophage migration inhibitory factor: a key cytokine and therapeutic target in colon cancer. Cytokine Growth Factor Rev 2015; 26:451-61. [PMID: 25882738 DOI: 10.1016/j.cytogfr.2015.03.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/25/2015] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF) was one of the first cytokines to be discovered, over 40 years ago. Since that time a burgeoning interest has developed in the role that MIF plays in both the regulation of normal physiology and the response to pathology. MIF is a pleotropic cytokine that functions to promote inflammation, drive cellular proliferation, inhibit apoptosis and regulate the migration and activation state of immune cells. These functions are particularly relevant for the development of cancer and it is notable that various solid tumours over express MIF. This includes tumours of the gastrointestinal tract and MIF appears to play a particularly prominent role in the development and progression of colonic adenocarcinoma. Here we review the role that MIF plays in colonic carcinogenesis through the promotion of colonic inflammation, as well as the progression of primary and metastatic colon cancer. The recent development of various antagonists and antibodies that inhibit MIF activity indicates that we may soon be able to classify MIF as a therapeutic target in colon cancer patients.
Collapse
Affiliation(s)
- A N Gordon-Weeks
- CRUK/MRC Gray Institute for Radiation Oncology & Biology, University of Oxford, UK.
| | - S Y Lim
- CRUK/MRC Gray Institute for Radiation Oncology & Biology, University of Oxford, UK
| | - A E Yuzhalin
- CRUK/MRC Gray Institute for Radiation Oncology & Biology, University of Oxford, UK
| | - K Jones
- CRUK/MRC Gray Institute for Radiation Oncology & Biology, University of Oxford, UK
| | - R Muschel
- CRUK/MRC Gray Institute for Radiation Oncology & Biology, University of Oxford, UK
| |
Collapse
|
20
|
Kawczyk-Krupka A, Bugaj AM, Latos W, Wawrzyniec K, Oleś P, Mertas A, Czuba Z, Król W, Sieroń-Stołtny K, Sieroń A. ALA-mediated photodynamic effect on apoptosis induction and secretion of macrophage migration inhibitory factor (MIF) and of monocyte chemotactic protein (MCP-1) by colon cancer cells in normoxia and in hypoxia-like conditions in vitro. Photodiagnosis Photodyn Ther 2015; 12:27-35. [DOI: 10.1016/j.pdpdt.2014.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
|
21
|
Das R, LaRose MI, Hergott CB, Leng L, Bucala R, Weiser JN. Macrophage migration inhibitory factor promotes clearance of pneumococcal colonization. THE JOURNAL OF IMMUNOLOGY 2014; 193:764-72. [PMID: 24928996 DOI: 10.4049/jimmunol.1400133] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human genetic polymorphisms associated with decreased expression of macrophage migration inhibitory factor (MIF) have been linked to the risk of community-acquired pneumonia. Because Streptococcus pneumoniae is the leading cause of community-acquired pneumonia and nasal carriage is a precursor to invasive disease, we explored the role of MIF in the clearance of pneumococcal colonization in a mouse model. MIF-deficient mice (Mif(-/-)) showed prolonged colonization with both avirulent (23F) and virulent (6A) pneumococcal serotypes compared with wild-type animals. Pneumococcal carriage led to both local upregulation of MIF expression and systemic increase of the cytokine. Delayed clearance in the Mif(-/-) mice was correlated with reduced numbers of macrophages in upper respiratory tract lavages as well as impaired upregulation of MCP-1/CCL2. We found that primary human monocyte-derived macrophages as well as THP-1 macrophages produced MIF upon pneumococcal infection in a pneumolysin-dependent manner. Pneumolysin-induced MIF production required its pore-forming activity and phosphorylation of p38-MAPK in macrophages, with sustained p38-MAPK phosphorylation abrogated in the setting of MIF deficiency. Challenge with pneumolysin-deficient bacteria demonstrated reduced MIF upregulation, decreased numbers of macrophages in the nasopharynx, and less effective clearance. Mif(-/-) mice also showed reduced Ab response to pneumococcal colonization and impaired ability to clear secondary carriage. Finally, local administration of MIF was able to restore bacterial clearance and macrophage accumulation in Mif(-/-) mice. Our work suggests that MIF is important for innate and adaptive immunity to pneumococcal colonization and could be a contributing factor in genetic differences in pneumococcal disease susceptibility.
Collapse
Affiliation(s)
- Rituparna Das
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| | - Meredith I LaRose
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| | - Christopher B Hergott
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| | - Lin Leng
- Department of Medicine, Yale School of Medicine, New Haven, CT 06510
| | - Richard Bucala
- Department of Medicine, Yale School of Medicine, New Haven, CT 06510
| | - Jeffrey N Weiser
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and
| |
Collapse
|
22
|
Sánchez-Zamora YI, Rodriguez-Sosa M. The role of MIF in type 1 and type 2 diabetes mellitus. J Diabetes Res 2014; 2014:804519. [PMID: 24527464 PMCID: PMC3910331 DOI: 10.1155/2014/804519] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/11/2013] [Indexed: 11/17/2022] Open
Abstract
Autoimmunity and chronic low-grade inflammation are hallmarks of diabetes mellitus type one (T1DM) and type two (T2DM), respectively. Both processes are orchestrated by inflammatory cytokines, including the macrophage migration inhibitory factor (MIF). To date, MIF has been implicated in both types of diabetes; therefore, understanding the role of MIF could affect our understanding of the autoimmune or inflammatory responses that influence diabetic pathology. This review highlights our current knowledge about the involvement of MIF in both types of diabetes in the clinical environment and in experimental disease models.
Collapse
Affiliation(s)
- Yuriko I. Sánchez-Zamora
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios No. 1, Los Reyes Iztacala, 54090 Tlalnepantla, MEX, Mexico
| | - Miriam Rodriguez-Sosa
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios No. 1, Los Reyes Iztacala, 54090 Tlalnepantla, MEX, Mexico
- *Miriam Rodriguez-Sosa:
| |
Collapse
|
23
|
Prencipe G, Auriti C, Inglese R, Gallusi G, Dotta A, De Benedetti F. The macrophage migration inhibitory factor -173G/C polymorphism is not significantly associated with necrotizing enterocolitis in preterm infants. J Pediatr Surg 2013; 48:1499-502. [PMID: 23895962 DOI: 10.1016/j.jpedsurg.2013.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 12/28/2012] [Accepted: 01/04/2013] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality among premature infants. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that has been implicated in the pathophysiology of inflammatory bowel diseases. The MIF promoter contains a functionally relevant single nucleotide polymorphism (SNP) G→C at position -173, with the MIF -173*C allele being associated with higher MIF expression in vitro and with higher MIF levels in vivo. The aim of this study was to investigate whether the G/C polymorphism at -173 of the MIF promoter is associated with the development of NEC. METHODS In this retrospective cohort study, 107 preterm infants (GA ≤ 32 weeks), of whom 41 had NEC (NEC Stage I n = 20, Stage II n = 3, Stage III n = 18) and 66 were not affected, were genotyped for the MIF -173 SNP. MIF genotyping was carried out by PCR and DHPLC. RESULTS We did not find significant differences in the prevalence of the -173G/C polymorphism and in the distribution of the -173 MIF genotype in infants with NEC compared to controls. Moreover, we did not observe an association between the polymorphism and mortality. CONCLUSIONS The polymorphism -173G/C of the MIF promoter does not appear to be of major importance in the pathophysiology of NEC in preterm infants.
Collapse
Affiliation(s)
- Giusi Prencipe
- Laboratory of Rheumatology, Bambino Gesù Children's Hospital, Roma, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Zhang H, Ma L, Dong LQ, Shu C, Xu JL. Association of the macrophage migration inhibitory factor gene--173G/C polymorphism with inflammatory bowel disease: a meta-analysis of 4296 subjects. Gene 2013; 526:228-31. [PMID: 23707797 DOI: 10.1016/j.gene.2013.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 05/01/2013] [Accepted: 05/06/2013] [Indexed: 12/26/2022]
Abstract
A variety of epidemiologic studies have focused on the association between macrophage migration inhibitory factor (MIF) gene--173G/C polymorphism and inflammatory bowel disease (IBD). However, results in different studies have been inconsistent. In order to derive a more precise estimation of the associations, we performed this meta-analysis and systematic searches of electronic databases PubMed and Web of Science (up to April 30, 2013). Based on our search criteria, a total of seven eligible studies concerning the MIF--173G/C polymorphism and IBD risk were included in the final meta-analysis, comprising 2162 IBD cases and 2134 controls. Significant association was found between MIF--173G/C polymorphism and the risk of IBD when all studies were pooled into the meta-analysis (for C allele vs. G allele: OR=1.25, 95% CI=1.12-1.41, p=0.000; for C/C vs. G/G: OR=1.71, 95% CI=1.23-2.39, p=0.002; for C/C+G/C vs. G/G: OR=1.24, 95% CI=1.09-1.42, p=0.002; for C/C vs. G/C+G/G: OR=1.67, 95% CI=1.20-2.33, p=0.002). Heterogeneity and publication bias did not exist in the overall comparisons. The present meta-analysis suggests an association between the MIF--173G/C polymorphism and IBD risk. However, due to few studies and the selection bias existed in some studies, the results should be interpreted with caution.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Neurosurgery, Air Force General Hospital of the Chinese PLA, 30 Fucheng Road, Haidian District, Beijing 100142, China
| | | | | | | | | |
Collapse
|
25
|
Lai X, Blazer-Yost BL, Clack JW, Fears SL, Mitra S, Ntim SA, Ringham HN, Witzmann FA. Protein expression profiles of intestinal epithelial co-cultures: effect of functionalised carbon nanotube exposure. ACTA ACUST UNITED AC 2013; 3. [PMID: 24228069 DOI: 10.1504/ijbnn.2013.054508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To assess the biological effects of low level, water dispersible, functionalised carbon nanotube (f-CNT) exposure in an in vitro model simulating the digestive tract, cellular protein expression was quantified and compared using label-free quantitative mass spectrometry (LFQMS). Co-cultured cells were exposed to well-characterised SWCNT-COOH, MWCNT-COOH, and MWCNT-PVP. The relative expression of 2,282 unique proteins was compared across the dose groups. 428 proteins were found to be differentially expressed. At the high dose, the extent of differential protein expression was CNT-specific and directly related to CNT colloidal stability. Cells responded to low level MWCNT-PVP exposure with three-fold greater differential expression. Bioinformatic analysis indicated significant and f-CNT-specific effects on relevant molecular and cellular functions and canonical pathways, with little overlap across f-CNT type and in the absence of overt toxicity.
Collapse
Affiliation(s)
- Xianyin Lai
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 1345 West 16th Street, Indianapolis IN 46202, USA,
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Clinicopathological significance of macrophage migration inhibitory factor and its relation with p53 in gastric cancer. J Gastrointest Cancer 2011; 42:5-10. [PMID: 20922580 DOI: 10.1007/s12029-010-9215-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIM Based on previous investigations, the progress of gastritis may lead to gastric carcinomas. In some epithelial tumors, macrophage migration inhibitory factor (MIF), which is an inflammatory cytokine may inactivate p53 and play a role in tumorigenesis process. We decided to evaluate clinicopathological significance of MIF expression and the relation between p53 and MIF expressions in gastric adenocarcinomas. METHODS Seventy-three consecutive cases of gastric adenocarcinomas, the tissue samples of which were available, were included in this study. Tissue sections were stained for MIF and p53 expression by immunohistochemistry and the expression was defined as positive (for more than 10%) and negative (for less than 10%) groups. Location of the tumor, histological subtypes, and grade of the tumor were determined by using routine H&E staining. Distant metastasis, lymph node involvement, and consequently the stage of tumor were specified. The patients' age and gender were obtained from their medical records. The relationship between expression of MIF and these variables was determined. RESULTS Overexpression of MIF was observed in the cytoplasm of cancer cells in 46.6% (34/73) of cases and nuclear immunostaining of p53 was observed in 37% (27/73) of cases. Expression of MIF was significantly correlated with the location of tumor, but this expression has no statistically significant correlation with variables including: age, gender histological subtypes, distant metastasis, and lymph node involvement, stage and grade of the tumor, and p53 tumor suppressor gene expression. CONCLUSIONS Our study suggests that MIF in gastric adenocarcinomas versus many other epithelial tumors cannot have a prominent role in tumor progress and inactivation of p53 tumor suppressor gene.
Collapse
|
27
|
The probiotic Escherichia coli Nissle 1917 reduces pathogen invasion and modulates cytokine expression in Caco-2 cells infected with Crohn's disease-associated E. coli LF82. Appl Environ Microbiol 2011; 77:2541-4. [PMID: 21317252 DOI: 10.1128/aem.01601-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Increased numbers of adherent invasive Escherichia coli (AIEC) have been found in Crohn's disease (CD) patients. In this report, we investigate the potential of the probiotic Escherichia coli Nissle 1917 (EcN) to reduce features associated with AIEC pathogenicity in an already established infection with AIEC reference strain LF82.
Collapse
|
28
|
Down-regulation of intestinal epithelial innate response by probiotic yeasts isolated from kefir. Int J Food Microbiol 2010; 140:102-8. [DOI: 10.1016/j.ijfoodmicro.2010.04.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 04/13/2010] [Accepted: 04/15/2010] [Indexed: 12/30/2022]
|
29
|
Dessein AF, Stechly L, Jonckheere N, Dumont P, Monté D, Leteurtre E, Truant S, Pruvot FR, Figeac M, Hebbar M, Lecellier CH, Lesuffleur T, Dessein R, Grard G, Dejonghe MJ, de Launoit Y, Furuichi Y, Prévost G, Porchet N, Gespach C, Huet G. Autocrine induction of invasive and metastatic phenotypes by the MIF-CXCR4 axis in drug-resistant human colon cancer cells. Cancer Res 2010; 70:4644-54. [PMID: 20460542 DOI: 10.1158/0008-5472.can-09-3828] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metastasis and drug resistance are major problems in cancer chemotherapy. The purpose of this work was to analyze the molecular mechanisms underlying the invasive potential of drug-resistant colon carcinoma cells. Cellular models included the parental HT-29 cell line and its drug-resistant derivatives selected after chronic treatment with either 5-fluorouracil, methotrexate, doxorubicin, or oxaliplatin. Drug-resistant invasive cells were compared with noninvasive cells using cDNA microarray, quantitative reverse transcription-PCR, flow cytometry, immunoblots, and ELISA. Functional and cellular signaling analyses were undertaken using pharmacologic inhibitors, function-blocking antibodies, and silencing by retrovirus-mediated RNA interference. 5-Fluorouracil- and methotrexate-resistant HT-29 cells expressing an invasive phenotype in collagen type I and a metastatic behavior in immunodeficient mice exhibited high expression of the chemokine receptor CXCR4. Macrophage migration-inhibitory factor (MIF) was identified as the critical autocrine CXCR4 ligand promoting invasion in drug-resistant colon carcinoma HT-29 cells. Silencing of CXCR4 and impairing the MIF-CXCR4 signaling pathways by ISO-1, pAb FL-115, AMD-3100, monoclonal antibody 12G5, and BIM-46187 abolished this aggressive phenotype. Induction of CXCR4 was associated with the upregulation of two genes encoding transcription factors previously shown to control CXCR4 expression (HIF-2alpha and ASCL2) and maintenance of intestinal stem cells (ASCL2). Enhanced CXCR4 expression was detected in liver metastases resected from patients with colon cancer treated by the standard FOLFOX regimen. Combination therapies targeting the CXCR4-MIF axis could potentially counteract the emergence of the invasive metastatic behavior in clonal derivatives of drug-resistant colon cancer cells.
Collapse
|
30
|
Wang F, Shen X, Guo X, Peng Y, Liu Y, Xu S, Yang J. Spinal macrophage migration inhibitory factor contributes to the pathogenesis of inflammatory hyperalgesia in rats. Pain 2009; 148:275-283. [PMID: 20005040 DOI: 10.1016/j.pain.2009.11.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 10/28/2009] [Accepted: 11/10/2009] [Indexed: 12/12/2022]
Abstract
Pro-inflammatory cytokine production after nociceptive stimuli is pivotal for hyperalgesia. As macrophage migration inhibitory factor (MIF), a pleiotropic cytokine produced mainly by nonneuronal tissue, has been involved in the regulation of neuronal functions, herein we examined the role for MIF in formalin-induced inflammatory pain model. MIF critically contributed to nociceptive behaviors following formalin injection. Specifically, spinal administration of a MIF inhibitor (ISO-1) prevented and reversed flinching responses in rats. Further examination showed that levels of both MIF and the MIF receptor CD74 were substantially increased within the ipsilateral spinal cord dorsal horn after formalin administration. Mechanistic studies revealed that MIF upregulated the expression of the spinal NMDA receptor subunit NR2B via the MAPK signaling pathway. Moreover, microglial cells were found to be the major source of spinal MIF after formalin administration by fluorescence colocalization. These data highlight spinal MIF plays a critical role in the pathogenesis of formalin-induced inflammatory pain and suggest MIF may be a potential target for therapy of such pathological condition.
Collapse
Affiliation(s)
- FuZhou Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, No. 22, Hankou Road, Nanjing 210093, Jiangsu, China Department of Anesthesiology, The Affiliated Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing 210004, Jiangsu, China The Institute of Pediatrics, The Affiliated Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing 210004, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Xia HHX, Yang Y, Chu KM, Gu Q, Zhang YY, He H, Wong WM, Leung SY, Yuen ST, Yuen MF, Chan AOO, Wong BCY. Serum macrophage migration-inhibitory factor as a diagnostic and prognostic biomarker for gastric cancer. Cancer 2009; 115:5441-9. [PMID: 19685530 DOI: 10.1002/cncr.24609] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND This study aimed to determine the potential diagnostic value of migration-inhibitory factor (MIF) for gastric cancer in patients presenting with dyspepsia and its prognostic value for gastric cancer. METHODS A cohort of 97 patients with histologically confirmed gastric adenocarcinoma and 222 patients with dyspepsia were recruited. Enzyme-linked immunosorbent assay was used to measure serum MIF and carcinoembryonic antigen (CEA). RESULTS The serum MIF concentrations were 6554.0 +/- 204.1 pg/mL and 1453.7 +/- 79.9 pg/mL, respectively, in gastric cancer patients and dyspeptic patients (P < .001). Serum MIF levels increased with the advancing gastric pathologies (P < .001). With the cutoff value of 3230 pg/mL, serum MIF had sensitivity, specificity, and accuracy of 83.5%, 92.3%, and 89.7%, respectively, in diagnosing gastric cancer, whereas the rates were 60.8%, 83.3%, and 76.5%, respectively, for serum CEA. Gastric cancer patients with serum MIF levels above 6600 pg/mL had a lower 5-year survival rate than those with serum MIF level below that level (P = .012). Higher serum CEA levels were also associated with poor survival. The prediction for 5-year survival was even better (P = .0001), using a combination of serum MIF and CEA. CONCLUSIONS Serum MIF level, which correlates with gastric MIF expression, is a better molecular marker than CEA in diagnosing gastric cancer in patients presenting with dyspepsia. A combination of serum MIF and CEA predicts 5-year survival better than the individual test.
Collapse
|
32
|
Lin S, Wang D, Iyer S, Ghaleb AM, Shim H, Yang VW, Chun J, Yun CC. The absence of LPA2 attenuates tumor formation in an experimental model of colitis-associated cancer. Gastroenterology 2009; 136:1711-20. [PMID: 19328876 PMCID: PMC2691721 DOI: 10.1053/j.gastro.2009.01.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 12/02/2008] [Accepted: 01/07/2009] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Chronic inflammation is a risk factor for colon cancer (CC). Lysophosphatidic acid (LPA), a naturally produced phospholipid, mediates multiple effects that are vital to disease process, including inflammation and cancer. The expression of LPA receptor 2 (LPA2) is up-regulated in several types of cancer, including ovarian and colon cancer, but the importance of LPA and LPA2 in the development and progression of CC is unclear. In this study, we sought to determine whether LPA and LPA2 regulate the progression of CC in vivo. METHODS We examined the potential role of LPA in CC progression by administering LPA to mice heterozygous for the adenomatous polyposis coli (Apc) allele. We determined the loss of LPA2 function in tumorigenesis in the colon by treating mice with genetic deletion of LPA2 (LPA2-/-) with azoxymethane and dextran sulfate sodium. RESULTS We found that LPA increased tumor incidence in Apc(min/+) mice. LPA2-/- mice showed reduced mucosal damage and fewer tumors than wild-type (WT) mice. Reduced epithelial cell proliferation and decreases in beta-catenin, Krüppel-like factor 5, and cyclooxygenase-2 expression were observed in LPA2-/- mice. Unlike WT mice, induction of monocyte chemoattractant protein-1 and macrophage migration inhibitory factor was significantly attenuated in LPA2-/- mice with reduced infiltration by macrophages. CONCLUSIONS These results show that LPA is capable of promoting tumorigenesis in the colon. The absence of LPA2 attenuates several effects that contribute to cancer progression in vivo, and, hence, the current study identifies LPA2 as an important modulator of CC.
Collapse
Affiliation(s)
- Songbai Lin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Man AL, Lodi F, Bertelli E, Regoli M, Pin C, Mulholland F, Satoskar AR, Taussig MJ, Nicoletti C. Macrophage migration inhibitory factor plays a role in the regulation of microfold (M) cell-mediated transport in the gut. THE JOURNAL OF IMMUNOLOGY 2008; 181:5673-80. [PMID: 18832726 DOI: 10.4049/jimmunol.181.8.5673] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been shown previously that certain bacteria rapidly (3 h) up-regulated in vivo microfold cell (M cell)-mediated transport of Ag across the follicle-associated epithelium of intestinal Peyer's patch. Our aim was to determine whether soluble mediators secreted following host-bacteria interaction were involved in this event. A combination of proteomics and immunohistochemical analyses was used to identify molecules produced in the gut in response to bacterial challenge in vivo; their effects were then tested on human intestinal epithelial cells in vitro. Macrophage migration inhibitory factor (MIF) was the only cytokine produced rapidly after in vivo bacterial challenge by CD11c(+) cells located beneath the M cell-rich area of the follicle-associated epithelium of the Peyer's patch. Subsequently, in vitro experiments conducted using human Caco-2 cells showed that, within hours, MIF induced the appearance of cells that showed temperature-dependent transport of microparticles and M cell-specific bacterium Vibrio cholerae, and acquired biochemical features of M cells. Furthermore, using an established in vitro human M cell model, we showed that anti-MIF Ab blocked Raji B cell-mediated conversion of Caco-2 cells into Ag-sampling cells. Finally, we report that MIF(-/-) mice, in contrast to wild-type mice, failed to show increased M cell-mediated transport following in vivo bacterial challenge. These data show that MIF plays a role in M cell-mediated transport, and cross-talk between bacteria, gut epithelium, and immune system is instrumental in regulating key functions of the gut, including M cell-mediated Ag sampling.
Collapse
Affiliation(s)
- Angela L Man
- Programme of Gastrointestinal Tract Biology and Health, Institute of Food Research, Norwich, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Willeke P, Gaubitz M, Schotte H, Maaser C, Domschke W, Schlüter B, Becker H. Increased serum levels of macrophage migration inhibitory factor in patients with primary Sjögren's syndrome. Arthritis Res Ther 2007; 9:R43. [PMID: 17470266 PMCID: PMC1906791 DOI: 10.1186/ar2182] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/16/2007] [Accepted: 04/30/2007] [Indexed: 11/21/2022] Open
Abstract
The objective of this study was to analyse levels of the proinflammatory cytokine macrophage migration inhibitory factor (MIF) in patients with primary Sjögren's syndrome (pSS) and to examine associations of MIF with clinical, serological and immunological variables. MIF was determined by ELISA in the sera of 76 patients with pSS. Further relevant cytokines (IL-1, IL-6, IL-10, IFN-γ and TNF-α) secreted by peripheral blood mononuclear cells (PBMC) were determined by ELISPOT assay. Lymphocytes and monocytes were examined flow-cytometrically for the expression of activation markers. Results were correlated with clinical and laboratory findings as well as with the HLA-DR genotype. Healthy age- and sex-matched volunteers served as controls. We found that MIF was increased in patients with pSS compared with healthy controls (p < 0.01). In particular, increased levels of MIF were associated with hypergammaglobulinemia. Further, we found a negative correlation of MIF levels with the number of IL-10-secreting PBMC in pSS patients (r = -0.389, p < 0.01). Our data indicate that MIF might participate in the pathogenesis of primary Sjögren's syndrome. MIF may contribute to B-cell hyperactivity indicated by hypergammaglobulinemia. The inverse relationship of IL-10 and MIF suggests that IL-10 works as an antagonist of MIF in pSS.
Collapse
Affiliation(s)
- Peter Willeke
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Markus Gaubitz
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Heiko Schotte
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Christian Maaser
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Wolfram Domschke
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Bernhard Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| | - Heidemarie Becker
- Department of Medicine B, Muenster University Hospital, Albert Schweitzer Strasse 33, 48129 Muenster, Germany
| |
Collapse
|
35
|
Hagemann T, Robinson SC, Thompson RG, Charles K, Kulbe H, Balkwill FR. Ovarian cancer cell-derived migration inhibitory factor enhances tumor growth, progression, and angiogenesis. Mol Cancer Ther 2007; 6:1993-2002. [PMID: 17620429 DOI: 10.1158/1535-7163.mct-07-0118] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In view of our previous findings that tumor cell-derived macrophage migration inhibitory factor (MIF) increased macrophage-mediated ovarian cancer cell invasiveness in vitro, we investigated the wider significance of ovarian cancer cell-derived MIF for tumor growth, metastasis, and angiogenesis. We found that MIF is expressed in borderline and malignant ovarian tumors, and active MIF is found in malignant ascitic fluid. We next investigated the expression and function of MIF in a syngeneic ovarian cancer model. Stable knockdown of MIF in the murine ovarian cancer cell line ID8 decreased in vivo tumor burden and overall survival. Tumors arising from MIF knockdown cells had decreased proliferation and significantly increased apoptosis. This was associated with an increased phosphorylation of p53 and reduced Akt phosphorylation. MIF knockdown led to a changed cytokine profile in the ascitic microenvironment; tumor necrosis factor-alpha, interleukin-6 (IL-6), and IL-10 expression were all significantly decreased. Accompanying this decrease in cytokine expression was a significant decrease in macrophage infiltration into ascites. Additionally, MIF knockdown reduced the expression of proangiogenic cytokines vascular endothelial growth factor and keratinocyte chemoattractant (KC) and reduced the amount of endothelial cells in the malignant ascites. We conclude that autocrine production of MIF by ovarian cancer cells stimulates other cytokines, chemokines, and angiogenic factors that may promote colonization of the peritoneum and neovascularization of tumor deposits.
Collapse
Affiliation(s)
- Thorsten Hagemann
- Queen Mary's Medical School, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | | | | | | | | | | |
Collapse
|
36
|
Cho Y, Jones BF, Vermeire JJ, Leng L, DiFedele L, Harrison LM, Xiong H, Kwong YKA, Chen Y, Bucala R, Lolis E, Cappello M. Structural and functional characterization of a secreted hookworm Macrophage Migration Inhibitory Factor (MIF) that interacts with the human MIF receptor CD74. J Biol Chem 2007; 282:23447-56. [PMID: 17567581 PMCID: PMC3707627 DOI: 10.1074/jbc.m702950200] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hookworms, parasitic nematodes that infect nearly one billion people worldwide, are a major cause of anemia and malnutrition. We hypothesize that hookworms actively manipulate the host immune response through the production of specific molecules designed to facilitate infection by larval stages and adult worm survival within the intestine. A full-length cDNA encoding a secreted orthologue of the human cytokine, Macrophage Migration Inhibitory Factor (MIF) has been cloned from the hookworm Ancylostoma ceylanicum. Elucidation of the three-dimensional crystal structure of recombinant AceMIF (rAceMIF) revealed an overall structural homology with significant differences in the tautomerase sites of the human and hookworm proteins. The relative bioactivities of human and hookworm MIF proteins were compared using in vitro assays of tautomerase activity, macrophage migration, and binding to MIF receptor CD74. The activity of rAceMIF was not inhibited by the ligand ISO-1, which was previously determined to be an inhibitor of the catalytic site of human MIF. These data define unique immunological, structural, and functional characteristics of AceMIF, thereby establishing the potential for selectively inhibiting the hookworm cytokine as a means of reducing parasite survival and disease pathogenesis.
Collapse
Affiliation(s)
- Yoonsang Cho
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Brian F. Jones
- Program in International Child Health and Department of Pediatrics , Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jon J. Vermeire
- Program in International Child Health and Department of Pediatrics , Yale University School of Medicine, New Haven, Connecticut 06520
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Lisa DiFedele
- Program in International Child Health and Department of Pediatrics , Yale University School of Medicine, New Haven, Connecticut 06520
| | - Lisa M. Harrison
- Program in International Child Health and Department of Pediatrics , Yale University School of Medicine, New Haven, Connecticut 06520
| | - Huabao Xiong
- Department of Pharmacology, Immunobiology Center, Mt. Sinai School of Medicine, New York, New York 10029
| | - Yuen-Kwan Amy Kwong
- Department of Pharmacology, Immunobiology Center, Mt. Sinai School of Medicine, New York, New York 10029
| | - Yibang Chen
- Department of Pharmacology, Immunobiology Center, Mt. Sinai School of Medicine, New York, New York 10029
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Elias Lolis
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
- To whom correspondence may be addressed: Dept. of Pharmacology, Yale University School of Medicine, New Haven, CT 06520.
| | - Michael Cappello
- Program in International Child Health and Department of Pediatrics , Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
37
|
Meléndez-López SG, Herdman S, Hirata K, Choi MH, Choe Y, Craik C, Caffrey CR, Hansell E, Chávez-Munguía B, Chen YT, Roush WR, McKerrow J, Eckmann L, Guo J, Stanley SL, Reed SL. Use of recombinant Entamoeba histolytica cysteine proteinase 1 to identify a potent inhibitor of amebic invasion in a human colonic model. EUKARYOTIC CELL 2007; 6:1130-6. [PMID: 17513563 PMCID: PMC1951106 DOI: 10.1128/ec.00094-07] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cysteine proteinases are key virulence factors of the protozoan parasite Entamoeba histolytica. We have shown that cysteine proteinases play a central role in tissue invasion and disruption of host defenses by digesting components of the extracellular matrix, immunoglobulins, complement, and cytokines. Analysis of the E. histolytica genome project has revealed more than 40 genes encoding cysteine proteinases. We have focused on E. histolytica cysteine proteinase 1 (EhCP1) because it is one of two cysteine proteinases unique to invasive E. histolytica and is highly expressed and released. Recombinant EhCP1 was expressed in Escherichia coli and refolded to an active enzyme with a pH optimum of 6.0. We used positional-scanning synthetic tetrapeptide combinatorial libraries to map the specificity of the P1 to P4 subsites of the active site cleft. Arginine was strongly preferred at P2, an unusual specificity among clan CA proteinases. A new vinyl sulfone inhibitor, WRR483, was synthesized based on this specificity to target EhCP1. Recombinant EhCP1 cleaved key components of the host immune system, C3, immunoglobulin G, and pro-interleukin-18, in a time- and dose-dependent manner. EhCP1 localized to large cytoplasmic vesicles, distinct from the sites of other proteinases. To gain insight into the role of secreted cysteine proteinases in amebic invasion, we tested the effect of the vinyl sulfone cysteine proteinase inhibitors K11777 and WRR483 on invasion of human colonic xenografts. The resultant dramatic inhibition of invasion by both inhibitors in this human colonic model of amebiasis strongly suggests a significant role of secreted amebic proteinases, such as EhCP1, in the pathogenesis of amebiasis.
Collapse
Affiliation(s)
- Samuel G Meléndez-López
- Department of Pathology, University of California, San Diego, San Diego, California 92103-8416, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Houdeau E, Moriez R, Leveque M, Salvador-Cartier C, Waget A, Leng L, Bueno L, Bucala R, Fioramonti J. Sex steroid regulation of macrophage migration inhibitory factor in normal and inflamed colon in the female rat. Gastroenterology 2007; 132:982-93. [PMID: 17324399 DOI: 10.1053/j.gastro.2006.12.028] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Accepted: 11/27/2006] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Sex steroids influence IBD symptoms. Macrophage migration inhibitory factor (MIF), a target of sex steroids in other inflammatory models, promotes interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha release in colitis. We investigated whether estradiol and progesterone influence MIF, IL-1beta, and TNF-alpha production in experimental colitis. METHODS Colonic MIF, IL-1beta, and TNF-alpha levels were measured in cyclic and ovariectomized rats, with or without estradiol benzoate (EB) or progesterone (P) replacement. MIF distribution was assessed by immunohistochemistry. Cytokines, myeloperoxidase activity, macroscopic damage, and plasma corticosterone were assessed 24 hours after intrarectal trinitrobenzene sulfonic acid (TNBS), with and without neutralizing anti-MIF antibody. Effects of EB and P on myeloperoxidase activity and MIF concentration were also assessed at 7 days in dextran sulfate sodium-induced colitis. RESULTS Basal IL-1beta and TNF-alpha contents did not fluctuate during the estrous cycle, while MIF concentrations increased from estrus (estrogen dominance) to metestrus (P dominance; P < .05). EB and P treatment mimicked these effects in ovariectomized rats, and similarly altered MIF immunostaining. Progesterone dominance aggravated TNBS colitis in comparison with estrogen. Progesterone enhanced TNBS-induced MIF (P < .001) and TNF-alpha (P < .01) production, while EB decreased MIF (P < .01) and IL-beta levels (P < .01). Anti-MIF antibody prevented P-mediated up-regulation of TNF-alpha, improved TNBS colitis, and enhanced plasma corticosterone. At 7 days after dextran sulfate sodium, EB decreased myeloperoxidase activity and MIF concentration, while P had no effect. CONCLUSIONS Estrogen decreases while progesterone increases MIF production in the female rat colon. Changes in basal MIF contents may affect colon susceptibility to inflammation, by modulating TNF-alpha and IL-1beta production during early stages of colitis.
Collapse
Affiliation(s)
- Eric Houdeau
- Neuro-Gastroenterology and Nutrition Unit, Institut National de la Recherche Agronomique, Toulouse, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dambacher J, Staudinger T, Seiderer J, Sisic Z, Schnitzler F, Pfennig S, Hofbauer K, Konrad A, Tillack C, Otte JM, Diebold J, Göke B, Ochsenkühn T, Lohse P, Brand S. Macrophage migration inhibitory factor (MIF) -173G/C promoter polymorphism influences upper gastrointestinal tract involvement and disease activity in patients with Crohn's disease. Inflamm Bowel Dis 2007; 13:71-82. [PMID: 17206642 DOI: 10.1002/ibd.20008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with increased expression in inflammatory bowel disease. The aim of the study was to analyze the role of the MIF -173G/C single nucleotide polymorphism in Crohn's disease (CD). METHODS Using restriction fragment length polymorphism analysis, genomic DNA of 198 patients with CD and 159 unrelated controls was analyzed for the -173G/C SNP in the MIF promoter region. Colonic MIF mRNA expression was measured by quantitative polymerase chain reaction (PCR), serum MIF levels by enzyme-linked immunosorbent assay (ELISA). RESULTS Thirty-six of the 146 G/G wildtype carriers (24.7%) but only 3 of the 45 G/C heterozygotes (6.7%) and only 1 of the C/C homozygotes (14.3%) were diagnosed with upper gastrointestinal tract involvement (P = 0.009, odds ratio [OR] = 0.22, 95% confidence interval [CI], 0.06-0.75 for wildtype versus hetero- and homozygous carriers). This result was confirmed in a second prospective study, in which all patients diagnosed with upper gastrointestinal involvement (n = 13) were G/G wildtype carriers (P = 0.01 versus controls). All patients (n = 12; 100%) with a Crohn's disease activity index (CDAI) > 300 were G/G wildtype carriers compared to only 65.6% wildtype carriers in the group with a CDAI < 150 (P = 0.016). MIF is expressed in the colonic mucosa of CD patients and intestinal epithelial cells but its mRNA expression does not correlate with the degree of inflammation and is not upregulated by proinflammatory cytokines. In CD, MIF serum levels are not influenced by the MIF -173G/C polymorphism. CONCLUSIONS The MIF -173G/C polymorphism appears to be a factor contributing to a particular CD phenotype characterized by protection against upper gastrointestinal tract involvement and severe disease activity.
Collapse
Affiliation(s)
- Julia Dambacher
- Department of Medicine II - Grosshadern, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Enjuanes L, Almazán F, Sola I, Zuñiga S. Biochemical aspects of coronavirus replication and virus-host interaction. Annu Rev Microbiol 2006; 60:211-30. [PMID: 16712436 DOI: 10.1146/annurev.micro.60.080805.142157] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infection by different coronaviruses (CoVs) causes alterations in the transcriptional and translational patterns, cell cycle, cytoskeleton, and apoptosis pathways of the host cells. In addition, CoV infection may cause inflammation, alter immune and stress responses, and modify the coagulation pathways. The balance between the up- and downregulated genes could explain the pathogenesis caused by these viruses. We review specific aspects of CoV-host interactions. CoV genome replication takes place in the cytoplasm in a membrane-protected microenvironment and may control the cell machinery by locating some of their proteins in the host cell nucleus. CoVs initiate translation by cap-dependent and cap-independent mechanisms. CoV transcription involves a discontinuous RNA synthesis (template switching) during the extension of a negative copy of the subgenomic mRNAs. The requirement for base-pairing during transcription has been formally demonstrated in arteriviruses and CoVs. CoV N proteins have RNA chaperone activity that may help initiate template switching. Both viral and cellular proteins are required for replication and transcription, and the role of selected proteins is addressed.
Collapse
Affiliation(s)
- Luis Enjuanes
- Department of Molecular and Cell Biology, CNB, CSIC, 28049 Madrid, Spain.
| | | | | | | |
Collapse
|
41
|
Lebiedz P, Heidemann J, Lugering A, Riedel S, Herbst H, Domschke W, Kucharzik T, Maaser C. Gastric epithelial expression of macrophage migration inhibitory factor is not altered by Helicobacter pylori infection in humans. Helicobacter 2006; 11:258-65. [PMID: 16882329 DOI: 10.1111/j.1523-5378.2006.00411.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent reports have shown an upregulation of macrophage migration inhibitory factor (MIF) during gastric ulcer development in a rat model and elevated counts of MIF-positive cells in biopsies from Helicobacter pylori-infected patients. H. pylori infection is a proven cofactor in humans causing gastritis and gastric ulcers. The aim of this study was to characterize MIF expression in human gastric epithelial cells in response to H. pylori. METHODS MIF mRNA and MIF protein expression was detected in human gastric epithelial cell lines after stimulation with proinflammatory cytokines or infection with H. pylori (cagA+/vacA+) using real-time reverse transcriptase-polymerase and enzyme-linked immunosorbent assay. Interleukin-8 secretion was measured as positive control. MIF mRNA and MIF protein expression was assessed in H. pylori-positive and -negative human gastric biopsy samples. RESULTS While interleukin-8 mRNA expression and interleukin-8 secretion were upregulated in gastric epithelial cells in vitro after H. pylori infection, no changes in MIF mRNA expression and MIF secretion could be detected. We found no significant differences in MIF expression in total RNA extracted from gastric biopsy tissue when comparing H. pylori-positive to control patients. Likewise, MIF protein expression in gastric epithelium was unaffected by H. pylori infection as compared to uninfected tissue. CONCLUSIONS While an increased MIF expression and positive effects of MIF blockade in ulcer healing have been shown in a rodent model and elevated numbers of MIF-positive cells have been found in H. pylori-infected human tissue, we herein could not confirm any differences in human gastric epithelial MIF expression and secretion after H. pylori infection in vitro and in vivo.
Collapse
Affiliation(s)
- Pia Lebiedz
- Department of Medicine B, University of Muenster, Albert-Schweitzer-Strasse 33, D-48129 Muenster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wu XY, Pan H, Huang P, Wu B, Jiang HJ, Mei L. Dinitrochlorobenzene-induced colitis and its correlations with neurogenic inflammation of gut in rats. Shijie Huaren Xiaohua Zazhi 2006; 14:2067-2072. [DOI: 10.11569/wcjd.v14.i21.2067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the immune mechanism of ulcerative colitis as well as the relationship between ulcerative colitis and enteric nervous system.
METHODS: Male Sprague Dawley rats were divided into 2, 4-dinitrochlorobenzene (DNCB) groups and ethanol (control) group. After sensitized by DNCB smeared on the abdominal skin, the rats were challenged with DNCB by enema or intrathecal injection. The weight, stool viscosity and hematochezia were observed and accumulated as the disease active index (DAI) score; the colon pathological score was achieved by macropathology and HE staining of section prepared for microscopy; and the activity of leukocyte migration inhibitory factor (LMIF) was determined by immunofluorescence staining in colon tissues.
RESULTS: In the rats treated with 4 and 8 g/L DNCB enema, the macropathological and microscopic scores were significantly higher than those in the controls (macropathological: 2.200 ± 0.416, 3.857 ± 0.143 vs 0.143 ± 0.143, P < 0.05; microscopic: 2.000 ± 0.471, 3.714 ± 0.184 vs 0.429 ± 0.297, P < 0.05). The colon tissue showed higher fluorescence intensity of LMIF in the rats treated with DNCB enema than that in the controls. In the rats intrathecally injected with 8 and 16 g/L DNCB, the DAI scores were significantly higher than those in the control rats (P < 0.05 and P <0.01). Colon HE staining showed mucosal edema in the rats intrathecally injected 4 g/L DNCB, infiltration of numerous inflammatory cells in those with 8 g/L DNCB, and mucosal erosion, necrosis, and ulceration in those with 16 g/L DNCB.
CONCLUSION: Colitis can be induced by DNCB enema or intrathecal injection in sensitized rat, which reflects a delayed type of hypersensitivity (DTH). The enteric nervous system and neuroimmune mediator LMIF play important roles in the DNCB-induced colitis.
Collapse
|
43
|
Skjolaas KA, Burkey TE, Dritz SS, Minton JE. Effects of Salmonella enterica serovars Typhimurium (ST) and Choleraesuis (SC) on chemokine and cytokine expression in swine ileum and jejunal epithelial cells. Vet Immunol Immunopathol 2006; 111:199-209. [PMID: 16473412 DOI: 10.1016/j.vetimm.2006.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 01/04/2006] [Indexed: 12/21/2022]
Abstract
The gastrointestinal epithelium represents a barrier to potentially invasive enteric pathogens, maintains a role in innate immune surveillance, and is a source of both chemokine and cytokine chemotactic mediators in response to bacterial invasion. In the current study, we evaluated cytokine and chemokine mediators known to regulate movement of macrophages (macrophage migration inhibitory factor; MIF), neutrophils (IL8), dendritic cells (CCL20), and epithelial remodeling (osteopontin; OPN) in response to invasive swine enteropathogens Salmonella enterica serovar Typhimurium (ST) or Choleraesuis (SC). For the in vivo experiment, weaned pigs served as uninfected controls (0 h) or were given 3 x 10(9) CFU ST orally. Pigs were sacrificed at 8, 24, 48, and 144 h after inoculation and total RNA was extracted from defined segments of proximal (PI) and distal (DI) ileum. Relative expression of MIF and OPN were not affected by ST. IL8 expression was increased numerically (P = 0.17 for the interaction term) at 24 and 144 h in the PI and these increases accounted for greater expression in the PI relative to the DI (P < 0.05). Relative expression of CCL20 was increased at 24 h after ST (P < 0.05). Next, we evaluated the time course of MIF, IL8, CCL20, and OPN mRNA expression induced by application of lipopolysaccharide (LPS), ST or SC in vitro using pig jejunal epithelial cells (IPEC-J2). Cells were grown to confluency on permeable membranes, and treated apically with LPS (10 ng/mL), ST or SC (10(8)/well). After 1 h, cells were washed to remove LPS or extracellular bacteria, and media containing gentamicin was added to kill remaining extracellular bacteria. Media and RNA were collected at 1.5, 3, and 6 h after treatment. MIF mRNA was not affected by LPS or bacterial treatment. Similarly, IL8 expression was not affected by LPS, but was increased by ST and SC relative to controls at 1.5 and 3 h post exposure (P < 0.05 for all comparisons). Treatment with SC increased CCL20 mRNA relative to controls at 3 h (P < 0.05), while ST increased CCL20 at 1.5, 3, and 6h with maximal expression at 6 h (P < 0.05 for all comparisons). ST and SC increased polarized IL8 secretion. Our data demonstrate that invasive bacterial pathogens in the pig gastrointestinal tract trigger upregulation of selected cytokine and chemokine mediators, but serovars of Salmonella elicited differing patterns of activation in vitro.
Collapse
Affiliation(s)
- K A Skjolaas
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506-0201, USA
| | | | | | | |
Collapse
|
44
|
Kimura K, Nagaki M, Nishihira J, Satake S, Kuwata K, Moriwaki H. Role of macrophage migration inhibitory factor in hepatitis B virus-specific cytotoxic-T-lymphocyte-induced liver injury. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2006; 13:415-9. [PMID: 16522786 PMCID: PMC1391960 DOI: 10.1128/cvi.13.3.415-419.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Macrophage migration inhibitory factor (MIF) plays a pivotal role in the development of various inflammatory diseases. Here, we found that anti-mouse MIF antibody treatment reduced liver injury and inflammatory cell infiltration into the liver after injection of antigen-specific cytotoxic T lymphocytes into hepatitis B virus transgenic mice.
Collapse
Affiliation(s)
- Kiminori Kimura
- First Department of Internal Medicine, Gifu University School of Medicine, 1-1 Yanagido, Gifu-shi Gifu 501-1194, Japan.
| | | | | | | | | | | |
Collapse
|
45
|
Becker H, Maaser C, Mickholz E, Dyong A, Domschke W, Gaubitz M. Relationship between serum levels of macrophage migration inhibitory factor and the activity of antineutrophil cytoplasmic antibody-associated vasculitides. Clin Rheumatol 2006; 25:368-72. [PMID: 16391884 DOI: 10.1007/s10067-005-0045-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Revised: 06/29/2005] [Accepted: 06/30/2005] [Indexed: 11/25/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a central proinflammatory cytokine that regulates innate and adaptive immune responses. To evaluate its role in primary vasculitides, we determined MIF by enzyme-linked immunoassay in the sera of patients with Wegener's granulomatosis (WG; n=26), microscopic polyangiitis (MPA; n=10), polyarteritis nodosa (PAN; n=9) and giant cell arteritis (GCA; n=11). Healthy controls (n=26) and patients with sarcoidosis (n=14) were studied in parallel. Serum levels of MIF were significantly higher in patients with WG (median 41.1, range 3.2-120 ng/ml) than those in healthy controls (6.0, 0.015-36.5 ng/ml; P<0.001) and in patients with sarcoidosis (13.8, 0.015-67.1 ng/ml; P<0.05). MIF values were higher in MPA patients (29.5, 9.9-69.4 ng/ml; P<0.01) in comparison with those in healthy controls. In particular, increased levels of MIF were associated with active disease as assessed by the Birmingham Vasculitis Activity Score. Sequential studies showed decreased levels of MIF after initiation of immunosuppressive therapy, with clinical improvement in WG and MPA patients. In contrast, serum levels of MIF were not significantly elevated in patients with PAN and GCA. The results suggest that MIF contributes to the inflammatory process and correlates with disease activity in antineutrophil cytoplasmic antibody-associated vasculitides.
Collapse
Affiliation(s)
- Heidemarie Becker
- Department of Medicine B, University of Muenster, Albert-Schweitzer-Strasse 33, 48129 Münster, Germany.
| | | | | | | | | | | |
Collapse
|
46
|
Ohkawara T, Takeda H, Miyashita K, Nishiwaki M, Nakayama T, Taniguchi M, Yoshiki T, Tanaka J, Takana J, Imamura M, Sugiyama T, Asaka M, Nishihira J. Regulation of Toll-like receptor 4 expression in mouse colon by macrophage migration inhibitory factor. Histochem Cell Biol 2005; 125:575-82. [PMID: 16283355 DOI: 10.1007/s00418-005-0092-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2005] [Indexed: 01/09/2023]
Abstract
Recent studies have indicated that macrophage migration inhibitory factor (MIF) and Toll-like receptor (TLR) play an important role in the regulation of innate immune responses. In this study, we investigated the effect of MIF on the expression of TLR4, a receptor that recognizes lipopolysaccharide, in colon using MIF-deficient mice. TLR4 mRNA expression in the colon tissues was determined by northern blot analysis. Western blot analysis and immunohistochemistry in the colon tissues were performed to evaluate the expression of TLR4 protein. The expressions of TLR4 mRNA and protein were remarkably down-regulated in colon tissues of MIF-deficient mice compared with wild-type mice and up-regulated by treatment with recombinant MIF. Immunohistochemical study revealed the presence of TLR4-positive staining in mononuclear cells in the lamina propria and intraepithelial mononuclear cells as well as weak staining in epithelial cells and crypts in colon tissues of wild-type mice. In contrast, MIF-deficient mice did not show TLR4-positive staining in the colonic mucosa. In MIF-deficient mice injected with recombinant mouse MIF (rMIF), TLR4-positive staining cells were observed in colon tissues similar to the findings in wild-type mice. Administration of dextran sulfate sodium (DSS) up-regulated the expression of TLR4 in the colons of WT mice but not in those of MIF-deficient mice. Furthermore, pretreatment with rMIF up-regulated the expression of TLR4 in response to DSS in MIF-deficient mice. Our results suggest that MIF affects the expression of TLR4 in mouse colon under both normal and colitic conditions.
Collapse
Affiliation(s)
- Tatsuya Ohkawara
- Department of Gastroenterology and Hematology, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, 060-8638, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wilson JM, Coletta PL, Cuthbert RJ, Scott N, MacLennan K, Hawcroft G, Leng L, Lubetsky JB, Jin KK, Lolis E, Medina F, Brieva JA, Poulsom R, Markham AF, Bucala R, Hull MA. Macrophage migration inhibitory factor promotes intestinal tumorigenesis. Gastroenterology 2005; 129:1485-503. [PMID: 16285950 DOI: 10.1053/j.gastro.2005.07.061] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Accepted: 07/21/2005] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS The cytokine macrophage migration inhibitory factor (MIF) is expressed throughout the human gastrointestinal tract. Recently, protumorigenic activity of MIF has been described in several cancer models. Therefore, we investigated the expression and function of MIF during the early stages of intestinal tumorigenesis. METHODS MIF messenger RNA, protein, and tautomerase activity were measured in normal intestinal mucosa and adenomas from patients with sporadic colorectal adenomas and in the adenomatous polyposis coli (Apc)Min/+ mouse model of intestinal tumorigenesis. MIF function was investigated by using VACO-235 human colorectal adenoma cells in vitro and by testing the effect of genetic deletion of Mif on ApcMin/+ mouse intestinal tumorigenesis. RESULTS MIF expression and tautomerase activity were increased in human and ApcMin/+ mouse intestinal adenomas compared with adjacent normal mucosa. Up-regulation of MIF occurred mainly in epithelial cells (associated with an increasing grade of dysplasia), but also in stromal plasma cells. Exogenous MIF inhibited apoptosis and promoted anchorage-independent growth of VACO-235 cells (maximal at 100 ng/mL). Homozygous deletion of Mif was associated with a reduction in the number and size of ApcMin/+ mouse adenomas (P = .025 for the difference in large [>7-mm] tumors) and decreased angiogenesis (43% decrease in mean tumor microvessel density), but there was no alteration in epithelial cell apoptosis or proliferation. CONCLUSIONS MIF expression is increased in sporadic human colorectal adenomas, and exogenous MIF drives tumorigenic behavior of epithelial cells in vitro. Mif also promotes intestinal tumorigenesis (predominantly via angiogenesis) in the ApcMin/+ mouse. Therefore, MIF is a potential colorectal cancer chemoprevention target.
Collapse
|
48
|
Ohkawara T, Takeda H, Nishihira J, Miyashita K, Nihiwaki M, Ishiguro Y, Takeda K, Akira S, Iwanaga T, Sugiyama T, Asaka M. Macrophage migration inhibitory factor contributes to the development of acute dextran sulphate sodium-induced colitis in Toll-like receptor 4 knockout mice. Clin Exp Immunol 2005; 141:412-21. [PMID: 16045730 PMCID: PMC1809451 DOI: 10.1111/j.1365-2249.2005.02877.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptor 4 (TLR4), which recognizes lipopolysaccharides, plays an important role in the innate immune response. In this study, we investigated the role of TLR4 in the development of experimental colitis with regard to the biological actions of macrophage migration inhibitory factor (MIF) using TLR4 null ((-/-)) mice. TLR4(-/-) mice were given 2% dextran sulphate sodium (DSS) in drinking water to induce colitis, which was clinically and histologically as severe as that seen in wild-type (WT) mice. The level of tumour necrosis factor (TNF)-alpha in colon tissues was increased in WT mice but unchanged in TLR4(-/-) mice. The level of myeloperoxidase (MPO) activity in colon tissues was increased by DSS administration in both TLR4(-/-) and WT mice. The expression of MIF was up-regulated in the colons of TLR4(-/-) mice with acute DSS-induced colitis. An anti-MIF antibody significantly suppressed colitis and elevation of matrix metalloproteinase-13 in TLR4(-/-) mice. The current results obtained from TLR4(-/-) mice provide evidence that MIF plays a critical role in the development of acute DSS-induced colitis.
Collapse
Affiliation(s)
- T Ohkawara
- Department of Gastroenterology and Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fontanella G, Bassan N, Vinuesa M. Sensitization increases esterase-positive macrophage number in appendix from an animal model of food allergy. Allergol Immunopathol (Madr) 2005; 33:277-81. [PMID: 16287547 DOI: 10.1157/13080931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Macrophages are mononuclear cells with phagocytic and antigen presenting properties. The role of macrophages in IgE-dependent allergic reactions and oral tolerance remains unclear. In previous works we demonstrated that ovalbumin (OVA)-sensitized rabbits present histopathological modifications of the mucosa in different regions of the digestive tract. The present study analyzes macrophage distribution and quantitative modifications in the cecal appendix of OVA-sensitized animals. METHODS Adult new Zealand rabbits were divided into two groups: G1 (non-sensitized normal controls) and G2 (rabbits sensitized to OVA twice by subcutaneous route, with aluminum hydroxide as adjuvant). The alpha-naphthyl esterase technique was used for macrophage detection. RESULTS Specific anti-OVA IgE was detected in sensitized animals by the PCA (passive cutaneous anaphylaxis) method. In 5 regions of the cecal appendix we observed a significant increase in the number of macrophages in sensitized animals (G2) versus the control group (G1). The observed sensitization-mediated increase in cells is probably related to enhanced recruitment of monocytes from peripheral blood towards the appendix. This process could be induced by chemical mediators, and demonstrates macrophage participation in local immune response during sensitization phenomena.
Collapse
Affiliation(s)
- G Fontanella
- Department of Histology and Embryology, Faculty of Medical Sciences, Rosario National University, Rosario, Argentina
| | | | | |
Collapse
|
50
|
Shun CT, Lin JT, Huang SP, Lin MT, Wu MS. Expression of macrophage migration inhibitory factor is associated with enhanced angiogenesis and advanced stage in gastric carcinomas. World J Gastroenterol 2005; 11:3767-71. [PMID: 15968736 PMCID: PMC4316032 DOI: 10.3748/wjg.v11.i24.3767] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Macrophage migration inhibitory factor (MIF) was reported to inactivate p53 and play an essential role in the growth and angiogenesis of tumors that arise at sites of chronic inflammation. Gastric inflammation is a prerequisite for the development of gastric carcinoma (GC), which has recently been linked to Helicobacter pylori (H pylori) infection. This study aimed to investigate clinicopathological significance of MIF expression in GCs.
METHODS: We selected 90 consecutive patients with GCs for investigation of the relation among MIF status, clinicopathological parameters, p53 expression and angiogenesis. MIF and p53 expression was assessed by immunohistochemistry as positive and negative groups. Tumor vascularity was evaluated by counting microvessel density on anti-CD34 stained sections. Expression status of MIF was correlated with determined clinicopathological data, p53 immunoreactivity and microvessel counts.
RESULTS: Strong immunostainings of MIF were observed in the cytoplasm of cancerous cells in 40% (36/90) of cases but not in normal or metaplastic epithelia. There was no statistically significant correlation between MIF expression and age, gender, H pylori infection, tumor location, histological subtypes, lymph node metastasis or p53 expression. Early GC less frequently overexpressed MIF as compared to advanced GCs (4/20 vs 32/70, P = 0.04). A remarkably increased microvessel count was noted in GCs with MIF expression than those without MIF expression (55.1±30.1 vs 31.3±28.8, P = 0.0001).
CONCLUSION: Our results suggest that expression of MIF may contribute to the progression and enhanced angiogenesis in a substantial portion of GCs.
Collapse
Affiliation(s)
- Chia-Tung Shun
- Department of Forensic Medicne and Pathology, National Taiwan University, Taipei, Taiwan, China
| | | | | | | | | |
Collapse
|