1
|
Yao J, Xu H, Ferrick-Kiddie EA, Nottingham RM, Wu DC, Ares M, Lambowitz AM. Human cells contain myriad excised linear intron RNAs with links to gene regulation and potential utility as biomarkers. PLoS Genet 2024; 20:e1011416. [PMID: 39325823 PMCID: PMC11460701 DOI: 10.1371/journal.pgen.1011416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/08/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
A previous study using Thermostable Group II Intron Reverse Transcriptase sequencing (TGIRT-seq) found human plasma contains short (≤300 nt) structured full-length excised linear intron (FLEXI) RNAs with potential to serve as blood-based biomarkers. Here, TGIRT-seq identified >9,000 different FLEXI RNAs in human cell lines, including relatively abundant FLEXIs with cell-type-specific expression patterns. Analysis of public CLIP-seq datasets identified 126 RNA-binding proteins (RBPs) that have binding sites within the region corresponding to the FLEXI or overlapping FLEXI splice sites in pre-mRNAs, including 53 RBPs with binding sites for ≥30 different FLEXIs. These included splicing factors, transcription factors, a chromatin remodeling protein, cellular growth regulators, and proteins with cytoplasmic functions. Analysis of ENCODE datasets identified subsets of these RBPs whose knockdown impacted FLEXI host gene mRNA levels or proximate alternative splicing, indicating functional interactions. Hierarchical clustering identified six subsets of RBPs whose FLEXI binding sites were co-enriched in six subsets of functionally related host genes: AGO1-4 and DICER, including but not limited to agotrons or mirtron pre-miRNAs; DKC1, NOLC1, SMNDC1, and AATF (Apoptosis Antagonizing Transcription Factor), including but not limited to snoRNA-encoding FLEXIs; two subsets of alternative splicing factors; and two subsets that included RBPs with cytoplasmic functions (e.g., LARP4, PABPC4, METAP2, and ZNF622) together with regulatory proteins. Cell fractionation experiments showed cytoplasmic enrichment of FLEXI RNAs with binding sites for RBPs with cytoplasmic functions. The subsets of host genes encoding FLEXIs with binding sites for different subsets of RBPs were co-enriched with non-FLEXI other short and long introns with binding sites for the same RBPs, suggesting overarching mechanisms for coordinately regulating expression of functionally related genes. Our findings identify FLEXIs as a previously unrecognized large class of cellular RNAs and provide a comprehensive roadmap for further analyzing their biological functions and the relationship of their RBPs to cellular regulatory mechanisms.
Collapse
Affiliation(s)
- Jun Yao
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| | - Hengyi Xu
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| | - Elizabeth A. Ferrick-Kiddie
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| | - Ryan M. Nottingham
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| | - Douglas C. Wu
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| | - Manuel Ares
- Department of Molecular, Cell, and Developmental Biology University of California, Santa Cruz, California, United States of America
| | - Alan M. Lambowitz
- Departments of Molecular Biosciences and Oncology University of Texas at Austin Austin, Texas, United States of America
| |
Collapse
|
2
|
Systematic analysis of different degrees of haemolysis on miRNA levels in serum and serum-derived extracellular vesicles from dogs. BMC Vet Res 2022; 18:355. [PMID: 36138476 PMCID: PMC9494854 DOI: 10.1186/s12917-022-03445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022] Open
Abstract
Background Circulating microRNAs (miRNAs) are described as promising non-invasive biomarkers for diagnostics and therapeutics. Human studies have shown that haemolysis occurring during blood collection or due to improper sample processing/storage significantly alters the miRNA content in plasma and serum. Nevertheless, no similar research has been performed in dogs so far. We therefore investigated the effects of different degrees of haemolysis on the levels of selected miRNAs in serum and serum-derived extracellular vesicles (EVs) from dogs, by inducing a controlled in vitro haemolysis experiment. Results The abundance of miR-16, miR-92a, miR-191, miR-451 and miR-486 was significantly sensitive to haemolysis in serum and serum-derived EVs, while other selected miRNAs were not influenced by haemolysis. Furthermore, we found that the abundance of some canine miRNAs differs from data reported in the human system. Conclusions Our results describe for the first time the impact of haemolysis on circulating miRNAs not only in whole serum, but also in serum-derived EVs from dogs. Hence, we provide novel data for further analyses in the discovery of canine circulating biomarkers. Our findings suggest that haemolysis should be carefully assessed to assure accuracy when investigating circulating miRNA in serum or plasma-based tests. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-022-03445-8.
Collapse
|
3
|
Xi D, Hofmann L, Alter T, Einspanier R, Bereswill S, Heimesaat MM, Gölz G, Sharbati S. The glycosyltransferase ST3GAL2 is regulated by miR-615-3p in the intestinal tract of Campylobacter jejuni infected mice. Gut Pathog 2021; 13:42. [PMID: 34183045 PMCID: PMC8240225 DOI: 10.1186/s13099-021-00437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/10/2021] [Indexed: 11/30/2022] Open
Abstract
Background Campylobacter jejuni (C. jejuni) infections are of increasing importance worldwide. As a typical mucosal pathogen, the interaction of C. jejuni with mucins is a prominent step in the colonisation of mucosal surfaces. Despite recent advances in understanding the interaction between bacterial pathogens and host mucins, the mechanisms of mucin glycosylation during intestinal C. jejuni infection remain largely unclear. This prompted us to identify relevant regulatory networks that are concerted by miRNAs and could play a role in the mucin modification and interaction. Results We firstly used a human intestinal in vitro model, in which we observed altered transcription of MUC2 and TFF3 upon C. jejuni NCTC 11168 infection. Using a combined approach consisting of in silico analysis together with in vitro expression analysis, we identified the conserved miRNAs miR-125a-5p and miR-615-3p associated with MUC2 and TFF3. Further pathway analyses showed that both miRNAs appear to regulate glycosyltransferases, which are related to the KEGG pathway ‘Mucin type O-glycan biosynthesis’. To validate the proposed interactions, we applied an in vivo approach utilising a well-established secondary abiotic IL-10−/− mouse model for infection with C. jejuni 81-176. In colonic tissue samples, we confirmed infection-dependent aberrant transcription of MUC2 and TFF3. Moreover, two predicted glycosyltransferases, the sialyltransferases ST3GAL1 and ST3GAL2, exhibited inversely correlated transcriptional levels compared to the expression of the identified miRNAs miR-125a-5p and miR-615-3p, respectively. In this study, we mainly focused on the interaction between miR-615-3p and ST3GAL2 and were able to demonstrate their molecular interaction using luciferase reporter assays and RNAi. Detection of ST3GAL2 in murine colonic tissue by immunofluorescence demonstrated reduced intensity after C. jejuni 81-176 infection and was thus consistent with the observations made above. Conclusions We report here for the first time the regulation of glycosyltransferases by miRNAs during murine infection with C. jejuni 81-176. Our data suggest that mucin type O-glycan biosynthesis is concerted by the interplay of miRNAs and glycosyltransferases, which could determine the shape of intestinal glycosylated proteins during infection. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-021-00437-1.
Collapse
Affiliation(s)
- De Xi
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lukas Hofmann
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stefan Bereswill
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Markus M Heimesaat
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Looney M, Lorenc R, Halushka MK, Karakousis PC. Key Macrophage Responses to Infection With Mycobacterium tuberculosis Are Co-Regulated by microRNAs and DNA Methylation. Front Immunol 2021; 12:685237. [PMID: 34140955 PMCID: PMC8204050 DOI: 10.3389/fimmu.2021.685237] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/13/2021] [Indexed: 02/03/2023] Open
Abstract
Tuberculosis (TB) is the leading cause of death from infection with a single bacterial pathogen. Host macrophages are the primary cell type infected with Mycobacterium tuberculosis (Mtb), the organism that causes TB. Macrophage response pathways are regulated by various factors, including microRNAs (miRNAs) and epigenetic changes that can shape the outcome of infection. Although dysregulation of both miRNAs and DNA methylation have been studied in the context of Mtb infection, studies have not yet investigated how these two processes may jointly co-regulate critical anti-TB pathways in primary human macrophages. In the current study, we integrated genome-wide analyses of miRNA abundance and DNA methylation status with mRNA transcriptomics in Mtb-infected primary human macrophages to decipher which macrophage functions may be subject to control by these two types of regulation. Using in vitro macrophage infection models and next generation sequencing, we found that miRNAs and methylation changes co-regulate important macrophage response processes, including immune cell activation, macrophage metabolism, and AMPK pathway signaling.
Collapse
Affiliation(s)
- Monika Looney
- Department of Medicine, Division of Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rachel Lorenc
- Department of Medicine, Division of Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C Karakousis
- Department of Medicine, Division of Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Riahi Rad Z, Riahi Rad Z, Goudarzi H, Goudarzi M, Mahmoudi M, Yasbolaghi Sharahi J, Hashemi A. MicroRNAs in the interaction between host-bacterial pathogens: A new perspective. J Cell Physiol 2021; 236:6249-6270. [PMID: 33599300 DOI: 10.1002/jcp.30333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Gene expression regulation plays a critical role in host-pathogen interactions, and RNAs function is essential in this process. miRNAs are small noncoding, endogenous RNA fragments that affect stability and/or translation of mRNAs, act as major posttranscriptional regulators of gene expression. miRNA is involved in regulating many biological or pathological processes through targeting specific mRNAs, including development, differentiation, apoptosis, cell cycle, cytoskeleton organization, and autophagy. Deregulated microRNA expression is associated with many types of diseases, including cancers, immune disturbances, and infection. miRNAs are a vital section of the host immune response to bacterial-made infection. Bacterial pathogens suppress host miRNA expression for their benefit, promoting survival, replication, and persistence. The role played through miRNAs in interaction with host-bacterial pathogen has been extensively studied in the past 10 years, and knowledge about these staggering molecules' function can clarify the complicated and ambiguous interactions of the host-bacterial pathogen. Here, we review how pathogens prevent the host miRNA expression. We briefly discuss emerging themes in this field, including their role as biomarkers in identifying bacterial infections, as part of the gut microbiota, on host miRNA expression.
Collapse
Affiliation(s)
- Zohreh Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Yasbolaghi Sharahi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Boonchuen P, Jaree P, Somboonviwat K, Somboonwiwat K. Regulation of shrimp prophenoloxidase activating system by lva-miR-4850 during bacterial infection. Sci Rep 2021; 11:3821. [PMID: 33589707 PMCID: PMC7884684 DOI: 10.1038/s41598-021-82881-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/15/2021] [Indexed: 01/31/2023] Open
Abstract
MicroRNAs (miRNAs) suppress gene expression and regulate biological processes. Following small RNA sequencing, shrimp hemocytes miRNAs differentially expressed in response to acute hepatopancreatic necrosis disease (AHPND) caused by Vibrio parahaemolyticus (VPAHPND) were discovered and some were confirmed by qRT-PCR. VPAHPND-responsive miRNAs were predicted to target several genes in various immune pathways. Among them, lva-miR-4850 is of interest because its predicted target mRNAs are two important genes of the proPO system; proPO2 (PO2) and proPO activating factor 2 (PPAF2). The expression of lva-miR-4850 was significantly decreased after VPAHPND infection, whereas those of the target mRNAs, PO2 and PPAF2, and PO activity were significantly upregulated. Introducing the lva-miR-4850 mimic into VPAHPND-infected shrimps caused a reduction in the PO2 and PPAF2 transcript levels and the PO activity, but significantly increased the number of bacteria in the VPAHPND targeted tissues. This result inferred that lva-miR-4850 plays a crucial role in regulating the proPO system via suppressing expression of PPAF2 and PO2. To fight against VPAHPND infection, shrimp downregulated lva-miR-4850 expression resulted in proPO activation.
Collapse
Affiliation(s)
- Pakpoom Boonchuen
- grid.7922.e0000 0001 0244 7875Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Phattarunda Jaree
- grid.10223.320000 0004 1937 0490Center of Applied Shrimp Research and Innovation, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom Thailand
| | - Kulwadee Somboonviwat
- grid.9723.f0000 0001 0944 049XFaculty of Engineering at Sriracha, Kasetsart University Sriracha Campus, Sriracha, Chonburi Thailand
| | - Kunlaya Somboonwiwat
- grid.7922.e0000 0001 0244 7875Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand ,grid.7922.e0000 0001 0244 7875Omics Science and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
7
|
Smith NC, Wajnberg G, Chacko S, Woldemariam NT, Lacroix J, Crapoulet N, Ayre DC, Lewis SM, Rise ML, Andreassen R, Christian SL. Characterization of miRNAs in Extracellular Vesicles Released From Atlantic Salmon Monocyte-Like and Macrophage-Like Cells. Front Immunol 2020; 11:587931. [PMID: 33262769 PMCID: PMC7686242 DOI: 10.3389/fimmu.2020.587931] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Cell-derived extracellular vesicles (EVs) participate in cell-cell communication via transfer of molecular cargo including genetic material like miRNAs. In mammals, it has previously been established that EV-mediated transfer of miRNAs can alter the development or function of immune cells, such as macrophages. Our previous research revealed that Atlantic salmon head kidney leukocytes (HKLs) change their morphology, phagocytic ability and miRNA profile from primarily “monocyte-like” at Day 1 to primarily “macrophage-like” at Day 5 of culture. Therefore, we aimed to characterize the miRNA cargo packaged in EVs released from these two cell populations. We successfully isolated EVs from Atlantic salmon HKL culture supernatants using the established Vn96 peptide-based pull-down. Isolation was validated using transmission electron microscopy, nanoparticle tracking analysis, and Western blotting. RNA-sequencing identified 19 differentially enriched (DE) miRNAs packaged in Day 1 versus Day 5 EVs. Several of the highly abundant miRNAs, including those that were DE (e.g. ssa-miR-146a, ssa-miR-155 and ssa-miR-731), were previously identified as DE in HKLs and are associated with macrophage differentiation and immune response in other species. Interestingly, the abundance relative of the miRNAs in EVs, including the most abundant miRNA (ssa-miR-125b), was different than the miRNA abundance in HKLs, indicating selective packaging of miRNAs in EVs. Further study of the miRNA cargo in EVs derived from fish immune cells will be an important next step in identifying EV biomarkers useful for evaluating immune cell function, fish health, or response to disease.
Collapse
Affiliation(s)
- Nicole C Smith
- Department of Ocean Sciences, Memorial University, St. John's, NL, Canada
| | | | - Simi Chacko
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Nardos T Woldemariam
- Department of Life Sciences and Health, OsloMet-Oslo Metropolitan University, Oslo, Norway
| | | | | | - D Craig Ayre
- Department of Molecular Sciences, University of Medicine and Health Sciences, Basseterre, Saint Kitts and Nevis
| | - Stephen M Lewis
- Atlantic Cancer Research Institute, Moncton, NB, Canada.,Department of Chemistry & Biochemistry, Université de Moncton, Moncton, NB, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Matthew L Rise
- Department of Ocean Sciences, Memorial University, St. John's, NL, Canada
| | - Rune Andreassen
- Department of Life Sciences and Health, OsloMet-Oslo Metropolitan University, Oslo, Norway
| | - Sherri L Christian
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada.,Department of Biochemistry, Memorial University, St. John's, NL, Canada
| |
Collapse
|
8
|
Aguilera-Rojas M, Sharbati S, Stein T, Einspanier R. Deregulation of miR-27a may contribute to canine fibroblast activation after coculture with a mast cell tumour cell line. FEBS Open Bio 2020; 10:802-816. [PMID: 32133790 PMCID: PMC7193169 DOI: 10.1002/2211-5463.12831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/27/2020] [Accepted: 03/03/2020] [Indexed: 12/26/2022] Open
Abstract
The tumour microenvironment comprises a diverse range of cells, including fibroblasts, immune cells and endothelial cells, along with extracellular matrix. In particular, fibroblasts are of significant interest as these cells are reprogrammed during tumorigenesis to become cancer‐associated fibroblasts (CAFs), which in turn support cancer cell growth. MicroRNAs (miRNAs) have been shown to be involved in this intercellular crosstalk in humans. To assess whether miRNAs are also involved in the activation of fibroblasts in dogs, we cocultured primary canine skin fibroblasts with the canine mast cell tumour cell line C2 directly or with C2‐derived exosomes, and measured differential abundance of selected miRNAs. Expression of the CAF markers alpha‐smooth muscle actin (ACTA2) and stanniocalcin 1 confirmed the activation of our fibroblasts after coculture. We show that fibroblasts displayed significant downregulation of miR‐27a and let‐7 family members. These changes correlated with significant upregulation of predicted target mRNAs. Furthermore, RNA interference knockdown of miR‐27a revealed that cyclin G1 (CCNG1) exhibited negative correlation at the mRNA and protein level, suggesting that CCNG1 is a target of miR‐27a in canine fibroblasts and involved in their activation. Importantly, miR‐27a knockdown itself resulted in fibroblast activation, as demonstrated by the formation of ACTA2 filaments. In addition, interleukin‐6 (IL‐6) was strongly induced in our fibroblasts when cocultured, indicating potential reciprocal signalling. Taken together, our findings are consistent with canine fibroblasts being reprogrammed into CAFs to further support cancer development and that downregulation of miR‐27a may play an important role in the tumour–microenvironment crosstalk.
Collapse
Affiliation(s)
- Matias Aguilera-Rojas
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Torsten Stein
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| |
Collapse
|
9
|
Aguilar C, Mano M, Eulalio A. Multifaceted Roles of microRNAs in Host-Bacterial Pathogen Interaction. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0002-2019. [PMID: 31152522 PMCID: PMC11026079 DOI: 10.1128/microbiolspec.bai-0002-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
10
|
Agliano F, Rathinam VA, Medvedev AE, Vanaja SK, Vella AT. Long Noncoding RNAs in Host-Pathogen Interactions. Trends Immunol 2019; 40:492-510. [PMID: 31053495 DOI: 10.1016/j.it.2019.04.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 02/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are key molecules that regulate gene expression in a variety of organisms. LncRNAs can drive different transcriptional and post-transcriptional events that impact cellular functions. Recent studies have identified many lncRNAs associated with immune cell development and activation; however, an understanding of their functional role in host immunity to infection is just emerging. Here, we provide a detailed and updated review of the functional roles of lncRNAs in regulating mammalian immune responses during host-pathogen interactions, because these functions may be either beneficial or detrimental to the host. With increased mechanistic insight into the roles of lncRNAs, it may be possible to design and/or improve lncRNA-based therapies to treat a variety of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Federica Agliano
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Vijay A Rathinam
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Andrei E Medvedev
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Sivapriya Kailasan Vanaja
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA.
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA.
| |
Collapse
|
11
|
Curtale G, Rubino M, Locati M. MicroRNAs as Molecular Switches in Macrophage Activation. Front Immunol 2019; 10:799. [PMID: 31057539 PMCID: PMC6478758 DOI: 10.3389/fimmu.2019.00799] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/26/2019] [Indexed: 12/25/2022] Open
Abstract
The efficacy of macrophage- mediated inflammatory response relies on the coordinated expression of key factors, which expression is finely regulated at both transcriptional and post-transcriptional level. Several studies have provided compelling evidence that microRNAs play pivotal roles in modulating macrophage activation, polarization, tissue infiltration, and resolution of inflammation. In this review, we highlight the essential molecular mechanisms underlying the different phases of inflammation that are targeted by microRNAs to inhibit or accelerate restoration to tissue integrity and homeostasis. We further review the impact of microRNA-dependent regulation of tumor-associated macrophages and the relative implication for tumor biology.
Collapse
Affiliation(s)
- Graziella Curtale
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Marcello Rubino
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| |
Collapse
|
12
|
Aguilar C, Mano M, Eulalio A. MicroRNAs at the Host-Bacteria Interface: Host Defense or Bacterial Offense. Trends Microbiol 2018; 27:206-218. [PMID: 30477908 DOI: 10.1016/j.tim.2018.10.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/17/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a class of small noncoding RNAs that act as major post-transcriptional regulators of gene expression. They are currently recognized for their important role in the intricate interaction between host and bacterial pathogens, either as part of the host immune response to neutralize infection, or as a molecular strategy employed by bacteria to hijack host pathways for their own benefit. Here, we summarize recent advances on the function of miRNAs during infection of mammalian hosts by bacterial pathogens, highlighting key cellular pathways. In addition, we discuss emerging themes in this field, including the participation of miRNAs in host-microbiota crosstalk and cell-to-cell communication.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
13
|
Ro Y, Jo G, Jung S, Lee E, Shin J, Lee J. Salmonella‑induced miR‑155 enhances necroptotic death in macrophage cells via targeting RIP1/3. Mol Med Rep 2018; 18:5133-5140. [DOI: 10.3892/mmr.2018.9525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/21/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Young‑Tae Ro
- Laboratory of Biochemistry, Graduate School of Medicine, Konkuk University, Chungju, Chungcheong 27478, Republic of Korea
| | - Guk‑Heui Jo
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Sun‑Ah Jung
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Eunjoo Lee
- Graduate School of East‑West Medical Science, Kyung Hee University, Yongin, Gyeonggi 17104, Republic of Korea
| | - Jongdae Shin
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| | - Joon Lee
- Laboratory of Cell Biology, Myunggok Eye Research Institute, Konyang University College of Medicine, Seoul 07301, Republic of Korea
| |
Collapse
|
14
|
Corsetti PP, de Almeida LA, Gonçalves ANA, Gomes MTR, Guimarães ES, Marques JT, Oliveira SC. miR-181a-5p Regulates TNF-α and miR-21a-5p Influences Gualynate-Binding Protein 5 and IL-10 Expression in Macrophages Affecting Host Control of Brucella abortus Infection. Front Immunol 2018; 9:1331. [PMID: 29942317 PMCID: PMC6004377 DOI: 10.3389/fimmu.2018.01331] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/29/2018] [Indexed: 12/13/2022] Open
Abstract
Brucella abortus is a Gram-negative intracellular bacterium that causes a worldwide zoonosis termed brucellosis, which is characterized as a debilitating infection with serious clinical manifestations leading to severe complications. In spite of great advances in studies involving host–B. abortus interactions, there are many gaps related to B. abortus modulation of the host immune response through regulatory mechanisms. Here, we deep sequenced small RNAs from bone marrow-derived macrophages infected with B. abortus, identifying 69 microRNAs (miRNAs) that were differentially expressed during infection. We further validated the expression of four upregulated and five downregulated miRNAs during infection in vitro that displayed the same profile in spleens from infected mice at 1, 3, or 6 days post-infection. Among these miRNAs, mmu-miR-181a-5p (upregulated) or mmu-miR-21a-5p (downregulated) were selected for further analysis. First, we determined that changes in the expression of both miRNAs induced by infection were dependent on the adaptor molecule MyD88. Furthermore, evaluating putative targets of mmu-miR-181a-5p, we demonstrated this miRNA negatively regulates TNF-α expression following Brucella infection. By contrast, miR-21a-5p targets included a negative regulator of IL-10, programmed cell death protein 4, and several guanylate-binding proteins (GBPs). As a result, during infection, miR-21a-5p led to upregulation of IL-10 expression and downregulation of GBP5 in macrophages infected with Brucella. Since GBP5 and IL-10 are important molecules involved in host control of Brucella infection, we decided to investigate the role of mmu-miR-21a-5p in bacterial replication in macrophages. We observed that treating macrophages with a mmu-miR-21a-5p mimic enhanced bacterial growth, whereas transfection of its inhibitor reduced Brucella load in macrophages. Taken together, the results indicate that downregulation of mmu-miR-21a-5p induced by infection increases GBP5 levels and decreases IL-10 expression thus contributing to bacterial control in host cells.
Collapse
Affiliation(s)
- Patrícia P Corsetti
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departmento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - Leonardo A de Almeida
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departmento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Brazil
| | - André Nicolau Aquime Gonçalves
- Laboratorio de Sorologia, Microbiologia e Biologia Molecular, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Marco Túlio R Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erika S Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - João T Marques
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Salvador, Brazil
| |
Collapse
|
15
|
Zur Bruegge J, Einspanier R, Sharbati S. A Long Journey Ahead: Long Non-coding RNAs in Bacterial Infections. Front Cell Infect Microbiol 2017; 7:95. [PMID: 28401065 PMCID: PMC5368183 DOI: 10.3389/fcimb.2017.00095] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/13/2017] [Indexed: 12/24/2022] Open
Abstract
Bacterial pathogens have coevolved with their hosts and acquired strategies to circumvent defense mechanisms of host cells. It was shown that bacteria interfere with the expression of mammalian microRNAs to modify immune signaling, autophagy, or the apoptotic machinery. Recently, a new class of regulatory RNAs, long non-coding RNAs (lncRNAs), was reported to have a pivotal role in the regulation of eukaryotic gene expression. A growing body of literature reports on specific involvement of lncRNAs in the host cell response toward bacterial infections. This mini review summarizes recent data that focuses on lncRNA function in host cells during bacterial infection and provides a perspective where future research in this regard may be going.
Collapse
Affiliation(s)
- Jennifer Zur Bruegge
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Ralf Einspanier
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Soroush Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| |
Collapse
|
16
|
Hanisch C, Sharbati J, Kutz-Lohroff B, Huber O, Einspanier R, Sharbati S. TFF3-dependent resistance of human colorectal adenocarcinoma cells HT-29/B6 to apoptosis is mediated by miR-491-5p regulation of lncRNA PRINS. Cell Death Discov 2017; 3:16106. [PMID: 28149533 PMCID: PMC5279457 DOI: 10.1038/cddiscovery.2016.106] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 12/21/2022] Open
Abstract
Tumour necrosis factor-α (TNF-α) is a double-edged cytokine associated with pathogenesis of inflammatory-related cancers being also able to induce cancer cell death. In the process of tumour development or metastasis, cancer cells can become resistant to TNF-α. In trefoil factor 3 (TFF3) overexpressing colorectal adenocarcinoma cells (HT-29/B6), we observed enhanced resistance against TNF-α/interferon gamma-induced apoptosis. TFF3 is a secreted small peptide that supports intestinal tissue repair but is also involved in intestinal tumour progression and scattering. We hypothesised that TFF3 rescues intestinal epithelial cancer cells from TNF-α-induced apoptosis by involving regulatory RNA networks. In silico-based expression analysis revealed TFF3-mediated regulation of selected microRNAs as well as long non-coding RNAs (lncRNAs), whereas miR-491-5p was identified to target the lncRNA ‘psoriasis susceptibility-related RNA gene induced by stress’ (PRINS). RNA interference-based gain- and loss-of-function experiments examined miR-491-PRINS axis to exert the TFF3-mediated phenotype. Chemical inhibition of selected pathways showed that phosphatidylinositol 3-kinase/AKT accounts for TFF3-mediated downregulation of miR-491-5p and accumulation of PRINS. Moreover, we showed that PRINS colocalises with PMAIP1 (NOXA) in nuclei of HT-29/B6 possessing inhibitory effects. Immunoprecipitation experiments proved molecular interaction of PMAIP1 with PRINS. Our study provides an insight into RNA regulatory networks that determine resistance of colorectal cancer cells to apoptosis.
Collapse
Affiliation(s)
- Carlos Hanisch
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Jutta Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany; Lise Meitner School of Science, Berlin, Germany
| | - Barbara Kutz-Lohroff
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Otmar Huber
- Institute of Biochemistry II, Jena University Hospital , Jena, Germany
| | - Ralf Einspanier
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Soroush Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| |
Collapse
|
17
|
Wu G, Qi Y, Liu X, Yang N, Xu G, Liu L, Li X. Cecal MicroRNAome response to Salmonella enterica serovar Enteritidis infection in White Leghorn Layer. BMC Genomics 2017; 18:77. [PMID: 28086873 PMCID: PMC5237128 DOI: 10.1186/s12864-016-3413-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/12/2016] [Indexed: 01/27/2023] Open
Abstract
Background Salmonella enterica serovar Enteritidis (SE) is a food-borne pathogen and of great threat to human health through consuming the contaminated poultry products. MicroRNAs (miRNAs) play an important role in different biological activities and have been shown to regulate the innate immunity in the bacterial infection. The objective of this study is to identify miRNAs associated with SE infection in laying chicken cecum. Results Average number of reads of three libraries constructed from infected and non-infected chickens was 12,476,156 and 10,866,976, respectively. There were 598 miRNAs including 194 potential novel miRNAs identified in which 37 miRNAs were significantly differentially expressed between infected and non-infected chickens. In total, 2897 unique target genes regulated by differentially expressed miRNAs were predicted, in which, 841 genes were uniquely regulated by up-regulated miRNAs (G1), 636 genes were uniquely regulated by down-regulated miRNAs (G2), and 1420 were co-regulated by both up and down- regulated miRNAs (G3). There were 118, 73 and 178 GO (Gene ontology) BP (Biological process) terms significantly enriched in G1, G2 and G3 groups, respectively. More immune-related GO BP terms than metabolism-related terms were found in G1. Expression of 12 immune-related genes of four differentially expressed miRNAs was detected through qRT-PCR. The regulatory direction of gga-miR-1416-5p, gga-miR-1662, and gga-miR-34a-5p were opposite with the target genes of TLR21, BCL10, TLR1LA, NOTCH2 and THBS1, respectively. Conclusion The miRNAs contribute to the response to SE infection at the onset of egg laying through regulating the homeostasis between metabolism and immunity. The gga-miR-125b-5p, gga-miR-34a-5p, gga-miR-1416-5p and gga-miR-1662 could play an important role in SE infection through regulating their target genes. The finding herein will pave the foundation for the studies of microRNA regulation in SE infection in laying hens. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3413-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guixian Wu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Yukai Qi
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Xiaoyi Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Ning Yang
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guiyun Xu
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Liying Liu
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, 271018, China.
| | - Xianyao Li
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, Shandong, 271018, China. .,Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Tai'an, Shandong, 271018, China.
| |
Collapse
|
18
|
Ahmed W, Zheng K, Liu ZF. Small Non-Coding RNAs: New Insights in Modulation of Host Immune Response by Intracellular Bacterial Pathogens. Front Immunol 2016; 7:431. [PMID: 27803700 PMCID: PMC5067535 DOI: 10.3389/fimmu.2016.00431] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 10/03/2016] [Indexed: 12/20/2022] Open
Abstract
Pathogenic bacteria possess intricate regulatory networks that temporally control the production of virulence factors and enable the bacteria to survive and proliferate within host cell. Small non-coding RNAs (sRNAs) have been identified as important regulators of gene expression in diverse biological contexts. Recent research has shown bacterial sRNAs involved in growth and development, cell proliferation, differentiation, metabolism, cell signaling, and immune response through regulating protein–protein interactions or via their ability to base pair with RNA and DNA. In this review, we provide a brief overview of mechanism of action employed by immune-related sRNAs, their known functions in immunity, and how they can be integrated into regulatory circuits that govern virulence, which will facilitate our understanding of pathogenesis and the development of novel, more effective therapeutic approaches to treat infections caused by intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Waqas Ahmed
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan , China
| | - Ke Zheng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan , China
| | - Zheng-Fei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University , Wuhan , China
| |
Collapse
|
19
|
Das K, Garnica O, Dhandayuthapani S. Modulation of Host miRNAs by Intracellular Bacterial Pathogens. Front Cell Infect Microbiol 2016; 6:79. [PMID: 27536558 PMCID: PMC4971075 DOI: 10.3389/fcimb.2016.00079] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate the expression of protein coding genes of viruses and eukaryotes at the post-transcriptional level. The eukaryotic genes regulated by miRNAs include those whose products are critical for biological processes such as cell proliferation, metabolic pathways, immune response, and development. It is now increasingly recognized that modulation of miRNAs associated with biological processes is one of the strategies adopted by bacterial pathogens to survive inside host cells. In this review, we present an overview of the recent findings on alterations of miRNAs in the host cells by facultative intracellular bacterial pathogens. In addition, we discuss how the altered miRNAs help in the survival of these pathogens in the intracellular environment.
Collapse
Affiliation(s)
| | | | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El PasoEl Paso, TX, USA
| |
Collapse
|
20
|
Zur Bruegge J, Backes C, Gölz G, Hemmrich-Stanisak G, Scharek-Tedin L, Franke A, Alter T, Einspanier R, Keller A, Sharbati S. MicroRNA Response of Primary Human Macrophages to Arcobacter Butzleri Infection. Eur J Microbiol Immunol (Bp) 2016; 6:99-108. [PMID: 27429792 PMCID: PMC4936332 DOI: 10.1556/1886.2016.00015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 01/02/2023] Open
Abstract
The role of microRNAs (miRNAs) in infectious diseases is becoming more and more apparent, and the use of miRNAs as a diagnostic tool and their therapeutic application has become the major focus of investigation. The aim of this study was to identify miRNAs involved in the immune signaling of macrophages in response to Arcobacter (A.) butzleri infection, an emerging foodborne pathogen causing gastroenteritis. Therefore, primary human macrophages were isolated and infected, and miRNA expression was studied by means of RNAseq. Analysis of the data revealed the expression of several miRNAs, which were previously associated with bacterial infections such as miR-155, miR-125, and miR-212. They were shown to play a key role in Toll-like receptor signaling where they act as fine-tuners to establish a balanced immune response. In addition, miRNAs which have yet not been identified during bacterial infections such as miR-3613, miR-2116, miR-671, miR-30d, and miR-629 were differentially regulated in A. butzleri-infected cells. Targets of these miRNAs accumulated in pathways such as apoptosis and endocytosis – processes that might be involved in A. butzleri pathogenesis. Our study contributes new findings about the interaction of A. butzleri with human innate immune cells helping to understand underlying regulatory mechanisms in macrophages during infection.
Collapse
Affiliation(s)
- Jennifer Zur Bruegge
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Christina Backes
- Chair for Clinical Bioinformatics, Saarland University , Germany
| | - Greta Gölz
- Institute of Food Hygiene, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | | | - Lydia Scharek-Tedin
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel , Germany
| | - Thomas Alter
- Institute of Food Hygiene, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University , Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| |
Collapse
|
21
|
Pawar K, Sharbati J, Einspanier R, Sharbati S. Mycobacterium bovis BCG Interferes with miR-3619-5p Control of Cathepsin S in the Process of Autophagy. Front Cell Infect Microbiol 2016; 6:27. [PMID: 27014637 PMCID: PMC4783571 DOI: 10.3389/fcimb.2016.00027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022] Open
Abstract
Main survival mechanism of pathogenic mycobacteria is to escape inimical phagolysosomal environment inside the macrophages. Many efforts have been made to unravel the molecular mechanisms behind this process. However, little is known about the involvement of microRNAs (miRNAs) in the regulation of phagolysosomal biosynthesis and maturation. Based on a bottom up approach, we searched for miRNAs that were involved in phagolysosomal processing events in the course of mycobacterial infection of macrophages. After infecting THP-1 derived macrophages with viable and heat killed Mycobacterium bovis BCG (BCG), early time points were identified after co-localization studies of the phagosomal marker protein LAMP1 and BCG. Differences in LAMP1 localization on the phagosomes of both groups were observed at 30 min and 4 h. After in silico based pre-selection of miRNAs, expression analysis at the identified time points revealed down-regulation of three miRNAs: miR-3619-5p, miR-637, and miR-324-3p. Consequently, most likely targets were predicted that were supposed to be mutually regulated by these three studied miRNAs. The lysosomal cysteine protease Cathepsin S (CTSS) and Rab11 family-interacting protein 4 (RAB11FIP4) were up-regulated and were considered to be connected to lysosomal trafficking and autophagy. Interaction studies verified the regulation of CTSS by miR-3619-5p. Down-regulation of CTSS by ectopic miR-3619-5p as well as its specific knockdown by siRNA affected the process of autophagy in THP-1 derived macrophages.
Collapse
Affiliation(s)
- Kamlesh Pawar
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Jutta Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Ralf Einspanier
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Soroush Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| |
Collapse
|
22
|
Yuhong J, Leilei T, Fuyun Z, Hongyang J, Xiaowen L, Liying Y, Lei Z, Jingrong M, Jinpeng Y. Identification and characterization of immune-related microRNAs in blunt snout bream, Megalobrama amblycephala. FISH & SHELLFISH IMMUNOLOGY 2016; 49:470-492. [PMID: 26773859 DOI: 10.1016/j.fsi.2015.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRNAs) play vital roles in diverse biological processes, including in immune response. Blunt snout bream (Megalobrama amblycephala) is a prevalent and important commercial endemic freshwater fish species in China's intensive polyculture systems. To identify immune-related miRNAs of M. amblycephala, two small RNA (sRNA) libraries from immune tissues with or without lipopolysaccharide (LPS) stimulation were constructed and sequenced using the high-throughput sequencing technology. Totally, 16,425,543 and 15,076,813 raw reads, corresponding to 14,156,755 and 13,445,869 clean reads, were obtained in the normal and infected libraries, respectively. A total of 324 miRNAs, including 218 known miRNAs and 106 putative novel miRNAs were identified by bioinformatic analysis. We analyzed differentially expressed miRNAs between two libraries using pairwise comparison. 113 (34.88%) miRNAs were found to be significantly differentially expressed between two libraries, with 63 (55.75%) exhibiting elevated expression in LPS stimulation sample. Thereinto, a number of known miRNAs were identified immune-related. Real-time quantitative PCR (RT-qPCR) were implemented for 12 miRNAs of two samples, and agreement was confirmed between the sequencing and RT-qPCR data. Target genes likely regulated by these differentially expressed miRNAs were predicted using computational prediction. The functional annotation of target genes by Gene Ontology enrichment (GO) and Kyoto Encyclopedia of Genes and Genomes pathway analysis (KEGG) indicated that a majority of differential miRNAs might involved in immune response. To our knowledge, this is the first comprehensive study of miRNAs in response to LPS stimulation in M. amblycephala, even in fish. These results deepened our understanding of the role of miRNAs in the intricate host's immune system, and should be useful to develop new control strategies for host immune defense against various bacterial invasions in M. amblycephala.
Collapse
Affiliation(s)
- Jiang Yuhong
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410017, China
| | - Tang Leilei
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410017, China
| | - Zhang Fuyun
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410017, China
| | - Jiang Hongyang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410017, China
| | - Liu Xiaowen
- Xiangya School of Medcine, Central South University, Changsha 410017, China
| | - Yang Liying
- Xiangya School of Medcine, Central South University, Changsha 410017, China
| | - Zhang Lei
- Xiangya School of Medcine, Central South University, Changsha 410017, China
| | - Mao Jingrong
- Xiangya School of Medcine, Central South University, Changsha 410017, China
| | - Yan Jinpeng
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha 410017, China.
| |
Collapse
|
23
|
Pawar K, Hanisch C, Palma Vera SE, Einspanier R, Sharbati S. Down regulated lncRNA MEG3 eliminates mycobacteria in macrophages via autophagy. Sci Rep 2016; 6:19416. [PMID: 26757825 PMCID: PMC4725832 DOI: 10.1038/srep19416] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
Small non-coding RNA play a major part in host response to bacterial agents. However, the role of long non-coding RNA (lncRNA) in this context remains unknown. LncRNA regulate gene expression by acting e.g. as transcriptional coactivators, RNA decoys or microRNA sponges. They control development, differentiation and cellular processes such as autophagy in disease conditions. Here, we provide an insight into the role of lncRNA in mycobacterial infections. Human macrophages were infected with Mycobacterium bovis BCG and lncRNA expression was studied early post infection. For this purpose, lncRNA with known immune related functions were preselected and a lncRNA specific RT-qPCR protocol was established. In addition to expression-based prediction of lncRNA function, we assessed strategies for thorough normalisation of lncRNA. Arrayed quantification showed infection-dependent repression of several lncRNA including MEG3. Pathway analysis linked MEG3 to mTOR and PI3K-AKT signalling pointing to regulation of autophagy. Accordingly, IFN-γ induced autophagy in infected macrophages resulted in sustained MEG3 down regulation and lack of IFN-γ allowed for counter regulation of MEG3 by viable M. bovis BCG. Knockdown of MEG3 in macrophages resulted in induction of autophagy and enhanced eradication of intracellular M. bovis BCG.
Collapse
Affiliation(s)
- Kamlesh Pawar
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Carlos Hanisch
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Sergio Eliseo Palma Vera
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| |
Collapse
|
24
|
Holla S, Balaji KN. Epigenetics and miRNA during bacteria-induced host immune responses. Epigenomics 2015; 7:1197-212. [PMID: 26585338 DOI: 10.2217/epi.15.75] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Various cellular processes including the pathogen-specific immune responses, host-pathogen interactions and the related evasion mechanisms rely on the ability of the immune cells to be reprogrammed accurately and in many cases instantaneously. In this context, the exact functions of epigenetic and miRNA-mediated regulation of genes, coupled with recent advent in techniques that aid such studies, make it an attractive field for research. Here, we review examples that involve the epigenetic and miRNA control of the host immune system during infection with bacteria. Interestingly, many pathogens utilize the epigenetic and miRNA machinery to modify and evade the host immune responses. Thus, we believe that global epigenetic and miRNA mapping of such host-pathogen interactions would provide key insights into their cellular functions and help to identify various determinants for therapeutic value.
Collapse
Affiliation(s)
- Sahana Holla
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | | |
Collapse
|
25
|
Saha B, Momen-Heravi F, Kodys K, Szabo G. MicroRNA Cargo of Extracellular Vesicles from Alcohol-exposed Monocytes Signals Naive Monocytes to Differentiate into M2 Macrophages. J Biol Chem 2015; 291:149-59. [PMID: 26527689 DOI: 10.1074/jbc.m115.694133] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Indexed: 12/19/2022] Open
Abstract
Membrane-coated extracellular vesicles (EVs) released by cells can serve as vehicles for delivery of biological materials and signals. Recently, we demonstrated that alcohol-treated hepatocytes cross-talk with immune cells via exosomes containing microRNA (miRNAs). Here, we hypothesized that alcohol-exposed monocytes can communicate with naive monocytes via EVs. We observed increased numbers of EVs, mostly exosomes, secreted by primary human monocytes and THP-1 monocytic cells in the presence of alcohol in a concentration- and time-dependent manner. EVs derived from alcohol-treated monocytes stimulated naive monocytes to polarize into M2 macrophages as indicated by increased surface expression of CD68 (macrophage marker), M2 markers (CD206 (mannose receptor) and CD163 (scavenger receptor)), secretion of IL-10, and TGFβ and increased phagocytic activity. miRNA profiling of the EVs derived from alcohol-treated THP-1 monocytes revealed high expression of the M2-polarizing miRNA, miR-27a. Treatment of naive monocytes with control EVs overexpressing miR-27a reproduced the effect of EVs from alcohol-treated monocytes on naive monocytes and induced M2 polarization, suggesting that the effect of alcohol EVs was mediated by miR-27a. We found that miR-27a modulated the process of phagocytosis by targeting CD206 expression on monocytes. Importantly, analysis of circulating EVs from plasma of alcoholic hepatitis patients revealed increased numbers of EVs that contained high levels of miR-27a as compared with healthy controls. Our results demonstrate the following: first, alcohol increases EV production in monocytes; second, alcohol-exposed monocytes communicate with naive monocytes via EVs; and third, miR-27a cargo in monocyte-derived EVs can program naive monocytes to polarize into M2 macrophages.
Collapse
Affiliation(s)
- Banishree Saha
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Fatemeh Momen-Heravi
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Karen Kodys
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Gyongyi Szabo
- From the Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
26
|
Zeng FR, Tang LJ, He Y, Garcia RC. An update on the role of miRNA-155 in pathogenic microbial infections. Microbes Infect 2015; 17:613-21. [PMID: 26072128 DOI: 10.1016/j.micinf.2015.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/28/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are evolutionarily conserved and naturally abundant molecules of single-stranded, non-coding RNA from ∼17 to 25 nucleotides long. MiRNAs act at post-transcriptional level either to suppress gene translation or to induce mRNA degradation, according to the degree of complementarity with their target sequences. MiR-155 is a typical representative of the miRNA family that plays a crucial role in cell differentiation and organism development. A number of studies have shown that miR-155 can not only regulate cell proliferation, apoptosis and lymphoma progression, but also plays an important part in various other physiological and pathological processes. For instance, it is involved in hematopoietic cell differentiation, cardiovascular disease, inflammation and immune responses. In recent years, the role of miR-155 in infectious diseases has attracted considerable attention. This review will highlight the participation of miR-155 in the responses to infections caused by different pathogens.
Collapse
Affiliation(s)
- Fu-Rong Zeng
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Li-Jun Tang
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China.
| | - Ye He
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - R C Garcia
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, 34012 Trieste, Italy
| |
Collapse
|
27
|
Saha B, Bruneau JC, Kodys K, Szabo G. Alcohol-induced miR-27a regulates differentiation and M2 macrophage polarization of normal human monocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:3079-87. [PMID: 25716995 PMCID: PMC4517579 DOI: 10.4049/jimmunol.1402190] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol abuse is a leading cause of liver disease characterized by liver inflammation, fatty liver, alcoholic hepatitis, or liver cirrhosis. Immunomodulatory effects of alcohol on monocytes and macrophages contribute to alcoholic liver disease. Alcohol use, an independent risk factor for progression of hepatitis C virus (HCV) infection-mediated liver disease, impairs host defense and alters cytokine production and monocyte/macrophage activation. We hypothesized that alcohol and HCV have synergistic effects on the phenotype and function of monocytes. Our data show that acute alcohol binge drinking in healthy volunteers results in increased frequency of CD16(+) and CD68(+) and M2-type (CD206(+), dendritic cell [DC]-SIGN(+)-expressing and IL-10-secreting) circulating CD14(+) monocytes. Expression of HCV-induced CD68 and M2 markers (CD206 and DC-SIGN) in normal monocytes was further enhanced in the presence of alcohol. The levels of microRNA (miR)-27a was significantly upregulated in monocytes cultured in the presence of alcohol or alcohol and HCV as compared with HCV alone. The functional role of miR-27a in macrophage polarization was demonstrated by transfecting monocytes with an miR-27a inhibitor that resulted in reduced alcohol- and HCV- mediated monocyte activation (CD14 and CD68 expression), polarization (CD206 and DC-SIGN expression), and IL-10 secretion. Overexpression of miR-27a in monocytes enhanced IL-10 secretion via activation of the ERK signaling pathway. We found that miR-27a promoted ERK phosphorylation by downregulating the expression of ERK inhibitor sprouty2 in monocytes. Thus, we identified that sprouty2 is a target of miR-27a in human monocytes. In summary, our study demonstrates the regulatory role of miR-27a in alcohol-induced monocyte activation and polarization.
Collapse
Affiliation(s)
- Banishree Saha
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Johanna C Bruneau
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Karen Kodys
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| |
Collapse
|
28
|
Fontemaggi G, Bellissimo T, Donzelli S, Iosue I, Benassi B, Bellotti G, Blandino G, Fazi F. Identification of post-transcriptional regulatory networks during myeloblast-to-monocyte differentiation transition. RNA Biol 2015; 12:690-700. [PMID: 25970317 PMCID: PMC4615388 DOI: 10.1080/15476286.2015.1044194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 01/24/2023] Open
Abstract
Treatment of leukemia cells with 1,25-dihydroxyvitamin D3 may overcome their differentiation block and lead to the transition from myeloblasts to monocytes. To identify microRNA-mRNA networks relevant for myeloid differentiation, we profiled the expression of mRNAs and microRNAs associated to the low- and high-density ribosomal fractions in leukemic cells and in their differentiated monocytic counterpart. Intersection between mRNAs shifted across the fractions after treatment with putative target genes of modulated microRNAs showed a series of molecular networks relevant for the monocyte cell fate determination, as for example the post-transcriptional regulation of the Polo-like kinase 1 (PLK1) by miR-22-3p and let-7e-5p.
Collapse
Key Words
- AGO2, argonaute 2
- AML
- AML, acute myeloid leukemia
- ECL methods, enhanced chemiluminescence methods
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GFP, green fluorescent protein
- HPCs, haematopoietic progenitor cells
- KPNA2, karyopherin α, 2
- NBT assay, nitroblue tetrazolium assay
- PLK1
- PLK1, polo-like kinase 1
- PMSF, phenylmethylsulfonyl fluoride
- RAB10, member RAS oncogene family 10
- RAB5C, member RAS oncogene family 5C
- RT-qPCR, quantitative reverse transcription polymerase chain reaction
- SF2A1, splicing factor 2A1
- TFs, transcription factors
- VitD3, 1,25-dihydroxyvitamin D3
- miRNAs, microRNAs
- microRNAs
- myeloid differentiation
- ribosomal/polysomal fractions
Collapse
Affiliation(s)
- Giulia Fontemaggi
- Translational Oncogenomics Unit; “Regina Elena” National Cancer Institute; Rome, Italy
| | - Teresa Bellissimo
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences; Section of Histology & Medical Embryology; Sapienza University of Rome; Rome, Italy
| | - Sara Donzelli
- Translational Oncogenomics Unit; “Regina Elena” National Cancer Institute; Rome, Italy
| | - Ilaria Iosue
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences; Section of Histology & Medical Embryology; Sapienza University of Rome; Rome, Italy
| | - Barbara Benassi
- Unit of Radiation Biology and Human Health; ENEA-Casaccia; Rome, Italy
| | | | - Giovanni Blandino
- Translational Oncogenomics Unit; “Regina Elena” National Cancer Institute; Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic, and Orthopedic Sciences; Section of Histology & Medical Embryology; Sapienza University of Rome; Rome, Italy
| |
Collapse
|
29
|
zur Bruegge J, Hanisch C, Einspanier R, Alter T, Gölz G, Sharbati S. Arcobacter butzleri induces a pro-inflammatory response in THP-1 derived macrophages and has limited ability for intracellular survival. Int J Med Microbiol 2014; 304:1209-17. [PMID: 25245281 DOI: 10.1016/j.ijmm.2014.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 08/29/2014] [Accepted: 08/30/2014] [Indexed: 02/03/2023] Open
Abstract
Recent case reports have identified Arcobacter (A.) butzleri to be another emerging pathogen of the family Campylobacteraceae causing foodborne diseases. However, little is known about its interaction with the human immune system. As macrophages act as first defense against bacterial infections, we studied for the first time the impact of A. butzleri on human macrophages using THP-1 derived macrophages as an in vitro infection model. Our investigations considered the inflammatory response, intracellular survival and activation of caspases as potential virulence mechanisms employed by A. butzleri. Induction of IL-1α, IL-1ß, IL-6, IL-8, IL-12ß and TNFα demonstrated a pro-inflammatory response of infected macrophages towards A. butzleri. gentamycin protection assays revealed the ability of A. butzleri strains to survive and resist the hostile environment of phagocytic immune cells for up to 22 h. Moreover, initial activation of intitiator- (CASP8) as well as effector caspases (CASP3/7) was observed without the onset of DNA damage, suggesting a potential counter regulation. Intriguingly, we recognized distinct strain specific differences in invasion and survival capabilities. This suggests the existence of isolate dependent phenotype variations and different virulence potentials as known for other intestinal pathogens such as Salmonella enterica ssp.
Collapse
Affiliation(s)
- Jennifer zur Bruegge
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany.
| | - Carlos Hanisch
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany.
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany.
| | - Thomas Alter
- Institute of Food Hygiene, Department of Veterinary Medicine, Freie Universität Berlin, Königsweg 69, 14163 Berlin, Germany.
| | - Greta Gölz
- Institute of Food Hygiene, Department of Veterinary Medicine, Freie Universität Berlin, Königsweg 69, 14163 Berlin, Germany.
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany.
| |
Collapse
|
30
|
MicroRNAs in the interaction between host and bacterial pathogens. FEBS Lett 2014; 588:4140-7. [PMID: 25128459 DOI: 10.1016/j.febslet.2014.08.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs with a central role in the post-transcriptional control of gene expression, that have been implicated in a wide-range of biological processes. Regulation of miRNA expression is increasingly recognized as a crucial part of the host response to infection by bacterial pathogens, as well as a novel molecular strategy exploited by bacteria to manipulate host cell pathways. Here, we review the current knowledge of bacterial pathogens that modulate host miRNA expression, focusing on mammalian host cells, and the implications of miRNA regulation on the outcome of infection. The emerging role of commensal bacteria, as part of the gut microbiota, on host miRNA expression in the presence or absence of bacterial pathogens is also discussed.
Collapse
|
31
|
MicroRNAs as Haematopoiesis Regulators. Adv Hematol 2013; 2013:695754. [PMID: 24454381 PMCID: PMC3884629 DOI: 10.1155/2013/695754] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 10/20/2013] [Accepted: 10/27/2013] [Indexed: 12/20/2022] Open
Abstract
The production of different types of blood cells including their formation, development, and differentiation is collectively known as haematopoiesis. Blood cells are divided into three lineages erythriod (erythrocytes), lymphoid (B and T cells), and myeloid (granulocytes, megakaryocytes, and macrophages). Haematopoiesis is a complex process regulated by several mechanisms including microRNAs (miRNAs). miRNAs are small RNAs which regulate the expression of a number of genes involved in commitment and differentiation of hematopoietic stem cells. Evidence shows that miRNAs play an important role in haematopoiesis; for example, myeloid and erythroid differentiation is blocked by the overexpression of miR-15a. miR-221, miR-222, and miR-24 inhibit the erythropoiesis, whereas miR-150 plays a role in B and T cell differentiation. miR-146 and miR-10a are downregulated in megakaryopoiesis. Aberrant expression of miRNAs was observed in hematological malignancies including chronic myelogenous leukemia, chronic lymphocytic leukemia, multiple myelomas, and B cell lymphomas. In this review we have focused on discussing the role of miRNA in haematopoiesis.
Collapse
|
32
|
Staedel C, Darfeuille F. MicroRNAs and bacterial infection. Cell Microbiol 2013; 15:1496-507. [PMID: 23795564 DOI: 10.1111/cmi.12159] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 12/13/2022]
Abstract
MicroRNAs, small non-coding RNAs expressed by eukaryotic cells, play pivotal roles in shaping cell differentiation and organism development. Deregulated microRNA expression is associated with several types of diseases including cancers, immune disorders and infection. Acting at the post-transcriptional level, miRNAs have expanded our understanding of the control of gene expression in regulatory networks involved in the adaptation to environmental situations such as biotic stress. It is increasingly clear that miRNAs are an important part of the host response to microbes. This review presents the current state of knowledge about the role of miRNAs in the response to both bacterial pathogens and commensal bacteria in human cells or animal experimental models. Some microRNAs, including miR-146, miR-155, miR-125, let-7 and miR-21, are commonly affected during bacterial infection and contribute to immune responses protecting the organism against overwhelmed inflammation. Cell-specific relationships between miRNAs and their targets are also engaged in the alterations induced by virulent bacteria in the proliferation/differentiation/apoptosis pathways of their host cells. In a separate role, miRNA modulation also represents a mechanism through which commensal bacteria impact the regulation of the barrier function and intestinal homeostasis.
Collapse
Affiliation(s)
- Cathy Staedel
- Univ. Bordeaux, ARNA Laboratory, F-33000, Bordeaux, France.
| | | |
Collapse
|
33
|
Tierney L, Kuchler K, Rizzetto L, Cavalieri D. Systems biology of host-fungus interactions: turning complexity into simplicity. Curr Opin Microbiol 2012; 15:440-6. [PMID: 22717554 PMCID: PMC3501689 DOI: 10.1016/j.mib.2012.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/24/2012] [Accepted: 05/01/2012] [Indexed: 12/15/2022]
Abstract
Modeling interactions between fungi and their hosts at the systems level requires a molecular understanding both of how the host orchestrates immune surveillance and tolerance, and how this activation, in turn, affects fungal adaptation and survival. The transition from the commensal to pathogenic state, and the co-evolution of fungal strains within their hosts, necessitates the molecular dissection of fungal traits responsible for these interactions. There has been a dramatic increase in publically available genome-wide resources addressing fungal pathophysiology and host–fungal immunology. The integration of these existing data and emerging large-scale technologies addressing host–pathogen interactions requires novel tools to connect genome-wide data sets and theoretical approaches with experimental validation so as to identify inherent and emerging properties of host–pathogen relationships and to obtain a holistic view of infectious processes. If successful, a better understanding of the immune response in health and microbial diseases will eventually emerge and pave the way for improved therapies.
Collapse
Affiliation(s)
- Lanay Tierney
- Medical University of Vienna, Christian Doppler Laboratory Infection Biology, Max F. Perutz Laboratories, A-1030 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Christian Doppler Laboratory Infection Biology, Max F. Perutz Laboratories, A-1030 Vienna, Austria
| | - Lisa Rizzetto
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Firenze, Italy
| | - Duccio Cavalieri
- Department of Preclinical and Clinical Pharmacology, University of Florence, 50139 Firenze, Italy
- Research and Innovation Centre, Edmund Mach Foundation, San Michele all’Adige, 38010, Trento, Italy
| |
Collapse
|