1
|
Maserumule C, Passemar C, Oh OSH, Hegyi K, Brown K, Weimann A, Dinan A, Davila S, Klapholz C, Bryant J, Verma D, Gadwa J, Krishnananthasivam S, Vongtongsalee K, Kendall E, Trelles A, Hibberd ML, Sanz J, Bertol J, Vázquez-Iniesta L, Andi K, Kumar SS, Ordway D, Prados-Rosales R, MacAry PA, Floto RA. Phagosomal RNA sensing through TLR8 controls susceptibility to tuberculosis. Cell Rep 2025; 44:115657. [PMID: 40338743 DOI: 10.1016/j.celrep.2025.115657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/13/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025] Open
Abstract
Genetic determinants of susceptibility to Mycobacterium tuberculosis (Mtb) remain poorly understood but could provide insights into critical pathways involved in infection, informing host-directed therapies and enabling risk stratification at individual and population levels. Through a genome-wide forward genetic screen, we identify Toll-like receptor 8 (TLR8) as a key regulator of intracellular killing of Mtb. Pharmacological TLR8 activation enhances the killing of phylogenetically diverse clinical isolates of drug-susceptible and multidrug-resistant Mtb by macrophages and during in vivo infection in mice. TLR8 is activated by phagosomal mycobacterial RNA released by extracellular membrane vesicles and enhances xenophagy-dependent Mtb killing. We find that the TLR8 variant M1V, common in Far Eastern populations, enhances intracellular killing of Mtb through preferential signal-dependent trafficking to phagosomes. TLR8 signaling may, therefore, both regulate susceptibility to tuberculosis and provide novel drug targets.
Collapse
Affiliation(s)
- Charlotte Maserumule
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Charlotte Passemar
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Victor Philip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Olivia S H Oh
- Department of Microbiology, The Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kriztina Hegyi
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Karen Brown
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Victor Philip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK; Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
| | - Aaron Weimann
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Victor Philip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK; Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
| | - Adam Dinan
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Victor Philip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK; Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
| | - Sonia Davila
- Infectious Disease Group, Genome Institute of Singapore, Singapore, Singapore; SingHealth Duke-NUS Institute of Precision Medicine, SingHealth Duke-NUS Genomic, Medicine Centre, Cardiovascular and Metabolic Disorder Program, Duke-NUS Medical, School, Singapore, Singapore
| | - Catherine Klapholz
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Josephine Bryant
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Jacob Gadwa
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Shivankari Krishnananthasivam
- Department of Microbiology, The Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kridakorn Vongtongsalee
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Edward Kendall
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Andres Trelles
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Martin L Hibberd
- Infectious Disease Group, Genome Institute of Singapore, Singapore, Singapore; London School of Hygiene and Tropical Medicine, London, UK
| | - Joaquín Sanz
- Institute for Bio-computation and Physics of Complex Systems BIFI, Department of Theoretical Physics, University of Zaragoza, Zaragoza, Spain
| | - Jorge Bertol
- Institute for Bio-computation and Physics of Complex Systems BIFI, Department of Theoretical Physics, University of Zaragoza, Zaragoza, Spain
| | - Lucia Vázquez-Iniesta
- Department of Preventive Medicine, Public Health and Microbiology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Kaliappan Andi
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - S Siva Kumar
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Diane Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Rafael Prados-Rosales
- Department of Preventive Medicine, Public Health and Microbiology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Paul A MacAry
- Department of Microbiology, The Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - R Andres Floto
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK; Victor Philip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK; Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK; Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Yang JY. miR-574-5p in epigenetic regulation and Toll-like receptor signaling. Cell Commun Signal 2024; 22:567. [PMID: 39593070 PMCID: PMC11600836 DOI: 10.1186/s12964-024-01934-x] [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: 09/12/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
miR-574-5p is an unusual microRNA (miRNA) that is often upregulated or downregulated following exposure to irradiation or toxic chemicals; bacterial, parasitic or viral infection; and a variety of other disease conditions. Canonically, miR-574-5p epigenetically regulates the expression of many messenger RNAs (mRNAs) through miRNA-mediated posttranscriptional regulation, thereby affecting cellular physiology or pathophysiology and contributing to the pathogenesis or progression of a variety of diseases. However, recent studies have established that in addition to serving as a fine-tuning repressor of gene expression, miR-574-5p also stimulates gene expression as an endogenous ligand for Toll-like receptor-8/7 (TLR8/7). Indeed, the binding of miR-574-5p to TLR8/7 triggers the TLR signaling pathway, leading to the induction of interferons, inflammatory cytokines and autoimmune signaling. These findings suggest that miR-574-5p is not only an important epigenetic regulator of gene expression, but also an important regulator of immune and inflammatory responses. Abnormal miR-574-5p-TLR8/7 signaling has been shown to be tightly associated with inflammation-related cancers and a number of autoimmune disorders. miR-574-5p can serve as a potential biomarker for many diseases. Most importantly, miR-574-5p is a promising therapeutic target for the treatment or prevention of human disorders, especially infectious diseases, cancers and autoimmune diseases.
Collapse
Affiliation(s)
- James Y Yang
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China.
- Wuhu Hospital of East China Normal University, Wuhu, 241000, Anhui, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China.
| |
Collapse
|
3
|
Kumaresan V, Hung CY, Hermann BP, Seshu J. Role of Dual Specificity Phosphatase 1 (DUSP1) in influencing inflammatory pathways in macrophages modulated by Borrelia burgdorferi lipoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624562. [PMID: 39605372 PMCID: PMC11601599 DOI: 10.1101/2024.11.20.624562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Borrelia burgdorferi (Bb), the spirochetal agent of Lyme disease, has a large array of lipoproteins that play a significant role in mediating host-pathogen interactions within ticks and vertebrates. Although there is substantial information on the effects of B. burgdorferi lipoproteins (BbLP) on immune modulatory pathways, the application of multi-omics methodologies to decode the transcriptional and proteomic patterns associated with host cell responses induced by lipoproteins in murine bone marrow-derived macrophages (BMDMs) has identified additional effectors and pathways. Single-cell RNA-Seq (scRNA-Seq) performed on BMDMs treated with various concentrations of borrelial lipoproteins revealed macrophage subsets within the BMDMs. Differential expression analysis showed that genes encoding various receptors, type I IFN-stimulated genes, signaling chemokines, and mitochondrial genes are altered in BMDMs in response to lipoproteins. Unbiased proteomics analysis of lysates of BMDMs treated with lipoproteins corroborated several of these findings. Notably, dual specificity phosphatase 1 (Dusp1) gene was upregulated during the early stages of BMDM exposure to BbLP. Pre-treatment with benzylidene-3-cyclohexylamino-1-indanone hydrochloride (BCI), an inhibitor of both DUSP1 and 6 prior to exposure to BbLP, demonstrated that DUSP1 negatively regulates NLRP3-mediated pro-inflammatory signaling and positively regulates the expression of interferon-stimulated genes and those encoding Ccl5, Il1b, and Cd274. Moreover, DUSP1, IkB kinase complex and MyD88 also modulate mitochondrial changes in BMDMs treated with borrelial lipoproteins. These findings advance the potential for exploiting DUSP1 as a therapeutic target to regulate host responses in reservoir hosts to limit survival of B. burgdorferi during its infectious cycle between ticks and mammalian hosts.
Collapse
Affiliation(s)
- Venkatesh Kumaresan
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| | - Chiung-Yu Hung
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX-78249
| | - J. Seshu
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX-78249
| |
Collapse
|
4
|
Nunes IV, Breitenbach L, Pawusch S, Eigenbrod T, Ananth S, Schad P, Fackler OT, Butter F, Dalpke AH, Chen LS. Bacterial RNA sensing by TLR8 requires RNase 6 processing and is inhibited by RNA 2'O-methylation. EMBO Rep 2024; 25:4674-4692. [PMID: 39363059 PMCID: PMC11549399 DOI: 10.1038/s44319-024-00281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
TLR8 senses single-stranded RNA (ssRNA) fragments, processed via cleavage by ribonuclease (RNase) T2 and RNase A family members. Processing by these RNases releases uridines and purine-terminated residues resulting in TLR8 activation. Monocytes show high expression of RNase 6, yet this RNase has not been analyzed for its physiological contribution to the recognition of bacterial RNA by TLR8. Here, we show a role for RNase 6 in TLR8 activation. BLaER1 cells, transdifferentiated into monocyte-like cells, as well as primary monocytes deficient for RNASE6 show a dampened TLR8-dependent response upon stimulation with isolated bacterial RNA (bRNA) and also upon infection with live bacteria. Pretreatment of bacterial RNA with recombinant RNase 6 generates fragments that induce TLR8 stimulation in RNase 6 knockout cells. 2'O-RNA methyl modification, when introduced at the first uridine in the UA dinucleotide, impairs processing by RNase 6 and dampens TLR8 stimulation. In summary, our data show that RNase 6 processes bacterial RNA and generates uridine-terminated breakdown products that activate TLR8.
Collapse
Affiliation(s)
- Ivanéia V Nunes
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| | - Luisa Breitenbach
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| | - Sarah Pawusch
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institute, Greifswald, Germany
| | - Tatjana Eigenbrod
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany
- Institute of Laboratory Medicine, SLK Clinics Heilbronn GmbH, 74078, Heilbronn, Germany
| | - Swetha Ananth
- Department of Infectious Diseases, Center for Integrative Infectious Disease Research (CIID), Integrative Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Paulina Schad
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Oliver T Fackler
- Department of Infectious Diseases, Center for Integrative Infectious Disease Research (CIID), Integrative Virology, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| | - Falk Butter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institute, Greifswald, Germany
| | - Alexander H Dalpke
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany.
- University Hospital Heidelberg, Heidelberg, Germany.
| | - Lan-Sun Chen
- Dept. of Infectious Diseases, Medical Microbiology and Hygiene, Medical Faculty, Heidelberg University, Heidelberg, Germany
- University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Berthold A, Lloyd VK. Changes in the Transcriptome and Long Non-Coding RNAs but Not the Methylome Occur in Human Cells Exposed to Borrelia burgdorferi. Genes (Basel) 2024; 15:1010. [PMID: 39202370 PMCID: PMC11353914 DOI: 10.3390/genes15081010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Lyme disease, caused by infection with members of the Lyme borreliosis group of Borrelia spirochete bacteria, is increasing in frequency and distribution worldwide. Epigenetic interactions between the mammalian host, tick, and bacterial pathogen are poorly understood. In this study, high-throughput next-generation sequencing (NGS) allowed for the in vitro study of the transcriptome, non-coding RNAs, and methylome in human host cells in response to Borrelia burgdorferi infection. We tested the effect of the Borrelia burgdorferi strain B31 on a human primary cell line (HUVEC) and an immortalized cell line (HEK-293) for 72 h, a long-duration time that might allow for epigenetic responses in the exposed human host cells. Differential gene expression was detected in both cell models in response to B. burgdorferi. More differentially expressed genes were found in HUVECs compared to HEK-293 cells. Borrelia burgdorferi exposure significantly induced genes in the interferon, in addition to cytokine and other immune response signaling in HUVECs. In HEK-293 cells, pre-NOTCH processing in Golgi was significantly downregulated in Borrelia-exposed cells. Other significantly altered gene expressions were found in genes involved in the extracellular matrix. No significant global methylation changes were detected in HUVECs or HEK-293 cells exposed to B. burgdorferi; however, two long non-coding RNAs and a pseudogene were deregulated in response to B. burgdorferi in HUVECs, suggesting that other epigenetic mechanisms may be initiated by infection.
Collapse
Affiliation(s)
| | - Vett K. Lloyd
- Department of Biology, Mount Allison University, Sackville, NB E4L 1G7, Canada;
| |
Collapse
|
6
|
Liu Y, He Y, Cao J, Lu H, Zou R, Zuo Z, Li R, Zhang Y, Sun J. Correlative analysis of transcriptome and proteome in Penaeus vannamei reveals key signaling pathways are involved in IFN-like antiviral regulation mediated by interferon regulatory factor (PvIRF). Int J Biol Macromol 2023; 253:127138. [PMID: 37776923 DOI: 10.1016/j.ijbiomac.2023.127138] [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: 07/01/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Interferon regulatory factors (IRFs) are crucial transcription factors that regulate interferon (IFN) induction in response to pathogen invasion. The regulatory mechanism of IRF has been well studied in vertebrates, but little has been known in arthropods. Therefore, in order to obtain new insights into the potential molecular mechanism of Peneaus vannamei IRF (PvIRF) in response to viral infection, comprehensive comparative analysis of the transcriptome and proteome profiles in shrimp infected with WSSV after knocking down PvIRF was conducted by using RNA sequencing (RNA-seq) and isobaric tags for relative and absolute quantification (iTRAQ). The sequence characterization, molecular functional evolution and 3D spatial structure of PvIRF were analyzed by using bioinformatics methods. PvIRF share the higher homology with different species in N-terminal end (containing DNA binding domain (DBD) including DNA sequence recognition sites and metal binding site) than that in C-terminal end. Within 4 IRF subfamilies of vertebrates, PvIRF had closer relationship with IRF1 subfamily. The DBD of PvIRF and C. gigas IRF1a were composed of α-helices and β-folds which was similar with the DBD structure of M. musculus IRF2. Interestingly, different from the five Tryptophan repeats highly homologous in the DBD of vertebrate IRF, the first and fifth tryptophans of PvIRF mutate to Phenylalanine and Leucine respectively, while the mutations were conserved among shrimp IRFs. RNAi knockdown of PvIRF gene by double-strand RNA could obviously promote the in vivo propagation of WSSV in shrimp and increase the mortality of WSSV-infected shrimp. It suggested that PvIRF was involved in inhibiting the replication of WSSV in shrimp. A total of 8787 transcripts and 2846 proteins were identified with significantly differential abundances in WSSV-infected shrimp after PvIRF knockdown, among which several immune-related members were identified and categorized into 10 groups according to their possible functions. Furthermore, the variation of expression profile from members of key signaling pathways involving JAK/STAT and Toll signaling pathway implied that they might participate IRF-mediated IFN-like regulation in shrimp. Correlative analyses indicated that 722 differentially expressed proteins (DEPs) shared the same expression profiles with their corresponding transcripts, including recognition-related proteins (CTLs and ITGs), chitin-binding proteins (peritrophin), and effectors (ALFs and SWD), while 401 DEPs with the opposite expression profiles across the two levels emphasized the critical role of post-transcriptional and post-translational modification. The results provide candidate signaling pathway including pivotal genes and proteins involved in the regulatory mechanism of interferon mediated by IRF on shrimp antiviral response. This is the first report in crustacean to explore the IFN-like antiviral regulation pathway mediated by IRF on the basis of transcriptome and proteomics correlative analysis, and will provide new ideas for further research on innate immune and defense mechanisms of crustacean.
Collapse
Affiliation(s)
- Yichen Liu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Yuxin He
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Jinlai Cao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Hangjia Lu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Ruifeng Zou
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Zhihan Zuo
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Ran Li
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Yichen Zhang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China.
| |
Collapse
|
7
|
Petnicki-Ocwieja T, McCarthy JE, Powale U, Langston PK, Helble JD, Hu LT. Borrelia burgdorferi initiates early transcriptional re-programming in macrophages that supports long-term suppression of inflammation. PLoS Pathog 2023; 19:e1011886. [PMID: 38157387 PMCID: PMC10783791 DOI: 10.1371/journal.ppat.1011886] [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: 08/05/2023] [Revised: 01/11/2024] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
Borrelia burgdorferi (Bb), the causative agent of Lyme disease, establishes a long-term infection and leads to disease manifestations that are the result of host immune responses to the pathogen. Inflammatory manifestations resolve spontaneously despite continued bacterial presence, suggesting inflammatory cells become less responsive over time. This is mimicked by in vitro repeated stimulations, resulting in tolerance, a phenotypic subset of innate immune memory. We performed comparative transcriptional analysis of macrophages in acute and memory states and identified sets of Tolerized, Hyper-Induced, Secondary-Induced and Hyper-Suppressed genes resulting from memory induction, revealing previously unexplored networks of genes affected by cellular re-programming. Tolerized gene families included inflammatory mediators and interferon related genes as would be predicted by the attenuation of inflammation over time. To better understand how cells mediate inflammatory hypo-responsiveness, we focused on genes that could mediate maintenance of suppression, such as Hyper-Induced genes which are up-regulated in memory states. These genes were notably enriched in stress pathways regulated by anti-inflammatory modulators. We examined one of the most highly expressed negative regulators of immune pathways during primary stimulation, Aconitate decarboxylase 1 (Acod1), and tested its effects during in vivo infection with Bb. As predicted by our in vitro model, we show its inflammation-suppressive downstream effects are sustained during in vivo long-term infection with Bb, with a specific role in Lyme carditis.
Collapse
Affiliation(s)
- Tanja Petnicki-Ocwieja
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Julie E. McCarthy
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Urmila Powale
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, United States of America
| | - P. Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, Massachusetts, United States of America
| | - Jennifer D. Helble
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
8
|
Farris LC, Torres-Odio S, Adams LG, West AP, Hyde JA. Borrelia burgdorferi Engages Mammalian Type I IFN Responses via the cGAS-STING Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1761-1770. [PMID: 37067290 PMCID: PMC10192154 DOI: 10.4049/jimmunol.2200354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 03/23/2023] [Indexed: 04/18/2023]
Abstract
Borrelia burgdorferi, the etiologic agent of Lyme disease, is a spirochete that modulates numerous host pathways to cause a chronic, multisystem inflammatory disease in humans. B. burgdorferi infection can lead to Lyme carditis, neurologic complications, and arthritis because of the ability of specific borrelial strains to disseminate, invade, and drive inflammation. B. burgdorferi elicits type I IFN (IFN-I) responses in mammalian cells and tissues that are associated with the development of severe arthritis or other Lyme-related complications. However, the innate immune sensors and signaling pathways controlling IFN-I induction remain unclear. In this study, we examined whether intracellular nucleic acid sensing is required for the induction of IFN-I to B. burgdorferi. Using fluorescence microscopy, we show that B. burgdorferi associates with mouse and human cells in culture, and we document that internalized spirochetes colocalize with the pattern recognition receptor cyclic GMP-AMP synthase (cGAS). Moreover, we report that IFN-I responses in mouse macrophages and murine embryonic fibroblasts are significantly attenuated in the absence of cGAS or its adaptor stimulator of IFN genes (STING), which function to sense and respond to intracellular DNA. Longitudinal in vivo tracking of bioluminescent B. burgdorferi revealed similar dissemination kinetics and borrelial load in C57BL/6J wild-type, cGAS-deficient, or STING-deficient mice. However, infection-associated tibiotarsal joint pathology and inflammation were modestly reduced in cGAS-deficient compared with wild-type mice. Collectively, these results indicate that the cGAS-STING pathway is a critical mediator of mammalian IFN-I signaling and innate immune responses to B. burgdorferi.
Collapse
Affiliation(s)
- Lauren C. Farris
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - L. Garry Adams
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - A. Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - Jenny A. Hyde
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| |
Collapse
|
9
|
Helicobacter pylori Infection Elicits Type I Interferon Response in Human Monocytes via Toll-Like Receptor 8 Signaling. J Immunol Res 2022; 2022:3861518. [PMID: 36317079 PMCID: PMC9617731 DOI: 10.1155/2022/3861518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/12/2022] [Accepted: 10/07/2022] [Indexed: 11/22/2022] Open
Abstract
Helicobacter pylori colonization and persistence could precede gastric adenocarcinoma. Elucidating immune recognition strategies of H. pylori is therefore imperative to curb chronic persistence in the human host. Toll-like receptor 7 (TLR7) and TLR8 are widely known as viral single-stranded RNA (ssRNA) sensors yet less studied in the bacteria context. Here, we investigated the involvement of these receptors in the immunity to H. pylori. Human THP-1 monocytic cells were infected with H. pylori, and the expression levels of human Toll-like receptors (TLRs) were examined. The roles of TLR7 and TLR8 in response to H. pylori infection were further investigated using receptor antagonists. Among all TLR transcripts examined, TLR8 exhibited the most prominent upregulation, followed by TLR7 in the THP-1 cells infected with H. pylori J99 or SS1 strains. H. pylori infection-mediated IFN-α and IFN-β transactivation was significantly abrogated by the TLR7/8 (but not TLR7) antagonist. Additionally, TLR7/8 antagonist treatment reduced H. pylori infection-mediated phosphorylation of interferon regulatory factor 7 (IRF7). Our study suggests a novel role of TLR8 signaling in host immunity against H. pylori through sensing live bacteria to elicit the production of type I interferon.
Collapse
|
10
|
Milillo MA, Velásquez LN, Barrionuevo P. Microbial RNA, the New PAMP of Many Faces. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.924719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Traditionally, pathogen-associated molecular patterns (PAMPs) were described as structural molecular motifs shared by different classes of microorganisms. However, it was later discovered that the innate immune system is also capable of distinguishing metabolically active microbes through the detection of a special class of viability-associated PAMPs (vita-PAMPs). Indeed, recognition of vita-PAMPs triggers an extra warning sign not provoked by dead bacteria. Bacterial RNA is classified as a vita-PAMP since it stops being synthesized once the microbes are eliminated. Most of the studies in the literature have focused on the pro-inflammatory capacity of bacterial RNA on macrophages, neutrophils, endothelial cells, among others. However, we, and other authors, have shown that microbial RNA also has down-modulatory properties. More specifically, bacterial RNA can reduce the surface expression of MHC class I and MHC class II on monocytes/macrophages and help evade CD8+ and CD4+ T cell-mediated immune surveillance. This phenomenon has been described for several different bacteria and parasites, suggesting that microbial RNA plays a significant immunoregulatory role in the context of many infectious processes. Thus, beyond the pro-inflammatory capacity of microbial RNA, it seems to be a crucial component in the intricate collection of immune evasion strategies. This review focuses on the different facets of the immune modulating capacity of microbial RNA.
Collapse
|
11
|
Oosting M, Brouwer M, Vrijmoeth HD, Pascual Domingo R, Greco A, ter Hofstede H, van den Bogaard EH, Schalkwijk J, Netea MG, Joosten LA. Borrelia burgdorferi is strong inducer of IFN-γ production by human primary NK cells. Cytokine 2022; 155:155895. [DOI: 10.1016/j.cyto.2022.155895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/05/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
|
12
|
Beasley EA, Pessôa-Pereira D, Scorza BM, Petersen CA. Epidemiologic, Clinical and Immunological Consequences of Co-Infections during Canine Leishmaniosis. Animals (Basel) 2021; 11:3206. [PMID: 34827938 PMCID: PMC8614518 DOI: 10.3390/ani11113206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/24/2022] Open
Abstract
Canine leishmaniosis (CanL) is a vector-borne, parasitic disease. CanL is endemic in the Mediterranean basin and South America but also found in Northern Africa, Asia, and the U.S. Regions with both competent sand fly vectors and L. infantum parasites are also endemic for additional infectious diseases that could cause co-infections in dogs. Growing evidence indicates that co-infections can impact immunologic responses and thus the clinical course of both CanL and the comorbid disease(s). The aim for this review is to summarize epidemiologic, clinical, and immunologic factors contributing to eight primary co-infections reported with CanL: Ehrlichia spp., Anaplasma spp., Borrelia spp., Babesia spp., Trypanosoma cruzi, Toxoplasma gondii, Dirofilaria immitis, Paracoccidioides braziliensis. Co-infection causes mechanistic differences in immunity which can alter diagnostics, therapeutic management, and prognosis of dogs with CanL. More research is needed to further explore immunomodulation during CanL co-infection(s) and their clinical impact.
Collapse
Affiliation(s)
- Erin A. Beasley
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Danielle Pessôa-Pereira
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Breanna M. Scorza
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| | - Christine A. Petersen
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA 52242, USA; (E.A.B.); (D.P.-P.); (B.M.S.)
- Center for Emerging Infectious Diseases, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
13
|
Akoolo L, Djokic V, Rocha SC, Parveen N. Pathogenesis of Borrelia burgdorferi and Babesia microti in TLR4-Competent and TLR4-dysfunctional C3H mice. Cell Microbiol 2021; 23:e13350. [PMID: 33938125 PMCID: PMC8459286 DOI: 10.1111/cmi.13350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/28/2022]
Abstract
Toll‐like receptors (TLRs) are a class of membrane‐spanning proteins of host cells. TLR2 and TLR4 are displayed on the surface of macrophages, neutrophils and dendritic cells and recognise structurally conserved microbial signatures defined as Pathogen associated molecular patterns (PAMPs). C3H mice are susceptible to tick‐borne pathogens; Lyme disease causing Borrelia burgdorferi that manifests arthritis and carditis and Apicomplexan protozoan, Babesia microti (Bm) that causes significant parasitemia associated with erythrocytopenia and haemoglobinuria. B. burgdorferi lacks typical TLR4 ligand lipopolysaccharides (LPS) and Bm TLR ligand(s) remain unknown. Only Borrelia lipoproteins that signal through TLR2 are established as PAMPs of these pathogens for TLR2/TLR4. Infection of C3H mice with each pathogen individually resulted in increase in the percentage of splenic B, T and FcR+ cells while their co‐infection significantly diminished levels of these cells and caused increased B. burgdorferi burden in the specific organs. The most pronounced inflammatory arthritis was observed in co‐infected C3H/HeJ mice. Parasitemia levels and kinetics of resolution of Bm in both mice strains were not significantly different. Transfected HEK293 cells showed pronounced signalling by B. burgdorferi through TLR2 and to some extent by TLR4 while Bm and infected erythrocytes did not show any response confirming our results in mice.
Collapse
Affiliation(s)
- Lavoisier Akoolo
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Vitomir Djokic
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Sandra C Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
14
|
Su Q, Chen Y, Wang B, Zhang Q, He H. Genetic characterizations of Toll-like receptors in the brown rat and their associations with pathogen infections. Integr Zool 2021; 17:879-889. [PMID: 34003606 DOI: 10.1111/1749-4877.12555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Toll-like receptors (TLRs) are important initiators of innate immune responses that target host-pathogen interactions. However, further research into the molecular characteristics of TLRs in wild populations is required, as well as how TLRs genetically influenced pathogen infections in the brown rat (Rattus norvegicus). Here, we explored the genetic characterization and evolution of 2 sensing nucleic acid TLRs (TLR7 and TLR8) and 2 sensing non-nucleic acid TLRs (TLR2 and TLR4) in the wild brown rat, and assessed their associations with 2 RNA viruses (Seoul hantavirus and rat hepatitis E virus (HEV)) and 2 bacteria (Leptospira and Bartonella). In these 4 TLRs, we discovered a total of 16 variants. Furthermore, TLR8 had high genetic diversity among 7 variants, while TLR2 had low genetic diversity with only 1 variant. According to selective pressure analyses, TLR4, TLR7, and TLR8 genes evolved under purifying selection. Interestingly, significant associations were found between 3 TLR8 variants and HEV infection, as well as 1 TLR2 variant and Bartonella infection. Overall, our findings provided a glimpse into the genetic characterization of TLRs in the brown rat, and further demonstrated that TLR2 and TLR8 genetic variations were related to Bartonella and HEV infection, respectively. Especially, TLR8 may be a good candidate immune gene for future research on molecular ecology and functional adaptation in wild populations.
Collapse
Affiliation(s)
- Qianqian Su
- National Research Center for Wildlife-Borne Diseases, Chinese Academy of Sciences, Institute of Zoology, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yi Chen
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of Sciences, Institute of Zoology, Beijing, China
| | - Bo Wang
- National Research Center for Wildlife-Borne Diseases, Chinese Academy of Sciences, Institute of Zoology, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qingxun Zhang
- National Research Center for Wildlife-Borne Diseases, Chinese Academy of Sciences, Institute of Zoology, Beijing, China
| | - Hongxuan He
- National Research Center for Wildlife-Borne Diseases, Chinese Academy of Sciences, Institute of Zoology, Beijing, China
| |
Collapse
|
15
|
Benjamin SJ, Hawley KL, Vera-Licona P, La Vake CJ, Cervantes JL, Ruan Y, Radolf JD, Salazar JC. Macrophage mediated recognition and clearance of Borrelia burgdorferi elicits MyD88-dependent and -independent phagosomal signals that contribute to phagocytosis and inflammation. BMC Immunol 2021; 22:32. [PMID: 34000990 PMCID: PMC8127205 DOI: 10.1186/s12865-021-00418-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Macrophages play prominent roles in bacteria recognition and clearance, including Borrelia burgdorferi (Bb), the Lyme disease spirochete. To elucidate mechanisms by which MyD88/TLR signaling enhances clearance of Bb by macrophages, we studied wildtype (WT) and MyD88-/- Bb-stimulated bone marrow-derived macrophages (BMDMs). RESULTS MyD88-/- BMDMs exhibit impaired uptake of spirochetes but comparable maturation of phagosomes following internalization of spirochetes. RNA-sequencing of infected WT and MyD88-/- BMDMs identified a large cohort of differentially expressed MyD88-dependent genes associated with re-organization of actin and cytoskeleton during phagocytosis along with several MyD88-independent chemokines involved in inflammatory cell recruitment. We computationally generated networks which identified several MyD88-dependent intermediate proteins (Rhoq and Cyfip1) that are known to mediate inflammation and phagocytosis respectively. CONCLUSION Our findings show that MyD88 signaling enhances, but is not required, for bacterial uptake or phagosomal maturation and provide mechanistic insights into how MyD88-mediated phagosomal signaling enhances Bb uptake and clearance.
Collapse
Affiliation(s)
- Sarah J Benjamin
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
| | - Kelly L Hawley
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA
| | - Paola Vera-Licona
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Center for Quantitative Medicine, UConn Health, Farmington, CT, 06030, USA
- Department of Cell Biology, UConn Health, Farmington, CT, 06030, USA
- Institute of Systems Genomics, UConn Health, Farmington, CT, 06030, USA
| | - Carson J La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
| | - Jorge L Cervantes
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA
- Present Address: Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, 79905, USA
| | - Yijun Ruan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Justin D Radolf
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
- Department of Medicine, UConn Health, Farmington, CT, 06030, USA
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, 06030, USA
- Department of Genetics and Genomic Sciences, UConn Health, Farmington, CT, 06030, USA
| | - Juan C Salazar
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA.
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA.
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA.
- Department of Medicine, UConn Health, Farmington, CT, 06030, USA.
- Division of Pediatric Infectious Diseases and Immunology, Connecticut Children's, 282 Washington Street, Hartford, CT, 06106, USA.
| |
Collapse
|
16
|
Barriales D, Martín-Ruiz I, Carreras-González A, Montesinos-Robledo M, Azkargorta M, Iloro I, Escobés I, Martín-Mateos T, Atondo E, Palacios A, Gonzalez-Lopez M, Bárcena L, Cortázar AR, Cabrera D, Peña-Cearra A, van Liempd SM, Falcón-Pérez JM, Pascual-Itoiz MA, Flores JM, Abecia L, Pellon A, Martínez-Chantar ML, Aransay AM, Pascual A, Elortza F, Berra E, Lavín JL, Rodríguez H, Anguita J. Borrelia burgdorferi infection induces long-term memory-like responses in macrophages with tissue-wide consequences in the heart. PLoS Biol 2021; 19:e3001062. [PMID: 33395408 PMCID: PMC7808612 DOI: 10.1371/journal.pbio.3001062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/14/2021] [Accepted: 12/22/2020] [Indexed: 11/19/2022] Open
Abstract
Lyme carditis is an extracutaneous manifestation of Lyme disease characterized by episodes of atrioventricular block of varying degrees and additional, less reported cardiomyopathies. The molecular changes associated with the response to Borrelia burgdorferi over the course of infection are poorly understood. Here, we identify broad transcriptomic and proteomic changes in the heart during infection that reveal a profound down-regulation of mitochondrial components. We also describe the long-term functional modulation of macrophages exposed to live bacteria, characterized by an augmented glycolytic output, increased spirochetal binding and internalization, and reduced inflammatory responses. In vitro, glycolysis inhibition reduces the production of tumor necrosis factor (TNF) by memory macrophages, whereas in vivo, it produces the reversion of the memory phenotype, the recovery of tissue mitochondrial components, and decreased inflammation and spirochetal burdens. These results show that B. burgdorferi induces long-term, memory-like responses in macrophages with tissue-wide consequences that are amenable to be manipulated in vivo. Lyme carditis is a manifestation of Lyme disease characterized by episodes of atrioventricular block and additional cardiomyopathies. This study describes the proteomic and transcriptomic changes in the heart upon infection with Borrelia burgdorferi, and identifies innate immune memory hallmarks specific to the response to the spirochete that are amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Itziar Martín-Ruiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ana Carreras-González
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Marta Montesinos-Robledo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Mikel Azkargorta
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Ibon Iloro
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Iraide Escobés
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Teresa Martín-Mateos
- Physiopathology of the Hypoxia-Signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Estibaliz Atondo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Laura Bárcena
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain
| | | | - Diana Cabrera
- Metabolomics Platform, CIC bioGUNE-BRTA, Derio, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Juan M. Falcón-Pérez
- Metabolomics Platform, CIC bioGUNE-BRTA, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Miguel A. Pascual-Itoiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Juana María Flores
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Complutense de Madrid, Madrid, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Aize Pellon
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Ana M. Aransay
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Felix Elortza
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Edurne Berra
- Physiopathology of the Hypoxia-Signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | | | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- * E-mail:
| |
Collapse
|
17
|
Bockenstedt LK, Wooten RM, Baumgarth N. Immune Response to Borrelia: Lessons from Lyme Disease Spirochetes. Curr Issues Mol Biol 2020; 42:145-190. [PMID: 33289684 PMCID: PMC10842262 DOI: 10.21775/cimb.042.145] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The mammalian host responds to infection with Borrelia spirochetes through a highly orchestrated immune defense involving innate and adaptive effector functions aimed toward limiting pathogen burdens, minimizing tissue injury, and preventing subsequent reinfection. The evolutionary adaptation of Borrelia spirochetes to their reservoir mammalian hosts may allow for its persistence despite this immune defense. This review summarizes our current understanding of the host immune response to B. burgdorferi sensu lato, the most widely studied Borrelia spp. and etiologic agent of Lyme borreliosis. Pertinent literature will be reviewed with emphasis on in vitro, ex vivo and animal studies that influenced our understanding of both the earliest responses to B. burgdorferi as it enters the mammalian host and those that evolve as spirochetes disseminate and establish infection in multiple tissues. Our focus is on the immune response of inbred mice, the most commonly studied animal model of B. burgdorferi infection and surrogate for one of this pathogen's principle natural reservoir hosts, the white-footed deer mouse. Comparison will be made to the immune responses of humans with Lyme borreliosis. Our goal is to provide an understanding of the dynamics of the mammalian immune response during infection with B. burgdorferi and its relation to the outcomes in reservoir (mouse) and non-reservoir (human) hosts.
Collapse
Affiliation(s)
- Linda K. Bockenstedt
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8031, USA
| | - R. Mark Wooten
- Department of Medical Microbiology and Immunology, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Nicole Baumgarth
- Center for Immunology and Infectious Diseases and Dept. Pathology, Microbiology and Immunology, University of California, Davis, Davis CA 95616, USA
| |
Collapse
|
18
|
Brouwer MAE, Jones-Warner W, Rahman S, Kerstholt M, Ferreira AV, Oosting M, Hooiveld GJ, Netea MG, Joosten LAB. B. burgdorferi sensu lato-induced inhibition of antigen presentation is mediated by RIP1 signaling resulting in impaired functional T cell responses towards Candida albicans. Ticks Tick Borne Dis 2020; 12:101611. [PMID: 33360386 DOI: 10.1016/j.ttbdis.2020.101611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Antigen presentation is a crucial innate immune cell function that instructs adaptive immune cells. Loss of this pathway severely impairs the development of adaptive immune responses. To investigate whether B. burgdorferi sensu lato. spirochetes modulate the induction of an effective immune response, primary human PBMCs were isolated from healthy volunteers and stimulated with B. burgdorferi s.l. Through cell entry, TNF receptor I, and RIP1 signaling cascades, B. burgdorferi s.l. strongly downregulated genes and proteins involved in antigen presentation, specifically HLA-DM, MHC class II and CD74. Antigen presentation proteins were distinctively inhibited in monocyte subsets, monocyte-derived macrophages, and dendritic cells. When compared to a range of other pathogens, B. burgdorferi s.l.-induced suppression of antigen presentation appears to be specific. Inhibition of antigen presentation interfered with T-cell recognition of B. burgdorferi s.l., and memory T-cell responses against Candidaalbicans. Re-stimulation of PBMCs with the commensal microbe C.albicans following B. burgdorferi s.l. exposure resulted in significantly reduced IFN-γ, IL-17 and IL-22 production. These findings may explain why patients with Lyme borreliosis develop delayed adaptive immune responses. Unravelling the mechanism of B. burgdorferi s.l.-induced inhibition of antigen presentation, via cell entry, TNF receptor I, and RIP1 signaling cascades, explains the difficulty to diagnose the disease based on serology and to obtain an effective vaccine against Lyme borreliosis.
Collapse
Affiliation(s)
- Michelle A E Brouwer
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - William Jones-Warner
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Shafaque Rahman
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mariska Kerstholt
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Marije Oosting
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Guido J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Zhong X, Lundberg M, Råberg L. Comparison of spleen transcriptomes of two wild rodent species reveals differences in the immune response against Borrelia afzelii. Ecol Evol 2020; 10:6421-6434. [PMID: 32724523 PMCID: PMC7381583 DOI: 10.1002/ece3.6377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/23/2020] [Accepted: 04/22/2020] [Indexed: 01/24/2023] Open
Abstract
Different host species often differ considerably in susceptibility to a given pathogen, but the causes of such differences are rarely known. The natural hosts of the tick-transmitted bacterium Borrelia afzelii, which is one of causative agents of Lyme borreliosis in humans, include a variety of small mammals like voles and mice. Previous studies have shown that B. afzelii-infected bank voles (Myodes glareolus) have about ten times higher bacterial load than infected yellow-necked mice (Apodemus flavicollis), indicating that these two species differ in resistance. In this study, we compared the immune response to B. afzelii infection in these host species by using RNA sequencing to quantify gene expression in spleen. Gene set enrichment analysis (GSEA) showed that several immune pathways were down-regulated in infected animals in both bank voles and yellow-necked mice. Moreover, IFNα response was up-regulated in B. afzelii-infected yellow-necked mice, while IL6 signaling and the complement pathway were down-regulated in infected bank voles; differences in regulation of these three pathways between bank voles and yellow-necked mice could thus contribute to the difference in resistance to B. afzelii between the species. This study provides knowledge of gene expression induced by a zoonotic pathogen in its natural host, and possible species-specific regulation of immune responses associated with resistance.
Collapse
Affiliation(s)
| | | | - Lars Råberg
- Department of BiologyLund UniversityLundSweden
| |
Collapse
|
20
|
Grabowski M, Bermudez M, Rudolf T, Šribar D, Varga P, Murgueitio MS, Wolber G, Rademann J, Weindl G. Identification and validation of a novel dual small-molecule TLR2/8 antagonist. Biochem Pharmacol 2020; 177:113957. [PMID: 32268138 DOI: 10.1016/j.bcp.2020.113957] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Toll-like receptor 2 (TLR2) and TLR8 are involved in the recognition of bacterial and viral components and are linked not only to protective antimicrobial immunity but also to inflammatory diseases. Recently, increasing attention has been paid to the receptor crosstalk between TLR2 and TLR8 to fine-tune innate immune responses. In this study, we report a novel dual TLR2/TLR8 antagonist, compound 24 that was developed by a modeling-guided synthesis approach. The modulator was optimized from the previously reported 1,3-benzothiazole derivative, compound 8. Compound 24 was pharmacologically characterized for the ability to inhibit TLR2- and TLR8-mediated responses in TLR-overexpressing reporter cells and THP-1 macrophages. The modulator showed high efficacy with IC50 values in the low micromolar range for both TLRs, selectivity towards other TLRs and low cytotoxicity. At TLR2, a slight predominance for the TLR2/1 heterodimer was found in reporter cells selectively expressing TLR2/1 or TLR2/6 heterodimers. Concentration ratio analysis in the presence of Pam3CSK4 or Pam2CSK4 indicated non-competitive antagonist behavior at hTLR2. In computational docking studies, a plausible alternative binding mode of compound 24 was predicted for both TLR2 and TLR8. Our results provide evidence that it is feasible to simultaneously and selectively target endosomal- and surface-located TLRs. We identified a small-molecule dual TLR2/8 antagonist that may serve as a valuable pharmacological tool to decipher the role of TLR2/8 co-signaling in inflammation.
Collapse
Affiliation(s)
- Maria Grabowski
- Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Marcel Bermudez
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Thomas Rudolf
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Dora Šribar
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Péter Varga
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Manuela S Murgueitio
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Gerhard Wolber
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Jörg Rademann
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Günther Weindl
- Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany; Section Pharmacology and Toxicology, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany.
| |
Collapse
|
21
|
Carreras-González A, Barriales D, Palacios A, Montesinos-Robledo M, Navasa N, Azkargorta M, Peña-Cearra A, Tomás-Cortázar J, Escobes I, Pascual-Itoiz MA, Hradiská J, Kopecký J, Gil-Carton D, Prados-Rosales R, Abecia L, Atondo E, Martín I, Pellón A, Elortza F, Rodríguez H, Anguita J. Regulation of macrophage activity by surface receptors contained within Borrelia burgdorferi-enriched phagosomal fractions. PLoS Pathog 2019; 15:e1008163. [PMID: 31738806 PMCID: PMC6886865 DOI: 10.1371/journal.ppat.1008163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/02/2019] [Accepted: 10/29/2019] [Indexed: 11/19/2022] Open
Abstract
Macrophages mediate the elimination of pathogens by phagocytosis resulting in the activation of specific signaling pathways that lead to the production of cytokines, chemokines and other factors. Borrelia burgdorferi, the causative agent of Lyme disease, causes a wide variety of pro-inflammatory symptoms. The proinflammatory capacity of macrophages is intimately related to the internalization of the spirochete. However, most receptors mediating this process are largely unknown. We have applied a multiomic approach, including the proteomic analysis of B. burgdorferi-containing phagosome-enriched fractions, to identify surface receptors that are involved in the phagocytic capacity of macrophages as well as their inflammatory output. Sucrose gradient protein fractions of human monocyte-derived macrophages exposed to B. burgdorferi contained the phagocytic receptor, CR3/CD14 highlighting the major role played by these proteins in spirochetal phagocytosis. Other proteins identified in these fractions include C-type lectins, scavenger receptors or Siglecs, of which some are directly involved in the interaction with the spirochete. We also identified the Fc gamma receptor pathway, including the binding receptor, CD64, as involved both in the phagocytosis of, and TNF induction in response to B. burgdorferi in the absence of antibodies. The common gamma chain, FcγR, mediates the phagocytosis of the spirochete, likely through Fc receptors and C-type lectins, in a process that involves Syk activation. Overall, these findings highlight the complex array of receptors involved in the phagocytic response of macrophages to B. burgdorferi. Macrophages eliminate infecting microorganisms through the concerted action of surface receptors and signaling molecules. As a consequence, these cells produce a series of soluble factors that participate in the inflammatory response during infections. The composition of the full complement of receptors that participate in the recognition and internalization of the causative agent of Lyme disease, Borrelia burgdorferi, is largely unknown. We have analyzed the protein composition of phagosomes containing B. burgdorferi from human macrophages and identified a series of surface proteins that may be involved in the process. Through the use of gene silencing techniques, we have determined the participation of several of these receptors both in the internalization of the bacterium and the subsequent inflammatory response. Among these, we have identified the Fc gamma receptor pathway as involved in this process in the absence of antibodies. We have also identified receptors that are directly involved in the attachment of B. burgdorferi, while others seem to have an accessory role in the internalization and/or induction of proinflammatory cytokines in response to the spirochete. These data clarify the complex array of interactions between macrophages and B. burgdorferi and shed light on the overall response to this infectious agent.
Collapse
Affiliation(s)
- Ana Carreras-González
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | | | - Nicolás Navasa
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Julen Tomás-Cortázar
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Iraide Escobes
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | | | - Jana Hradiská
- Faculty of Science, University of South Bohemia, Branišovská, České Budějovice, Czech Republic
| | - Jan Kopecký
- Faculty of Science, University of South Bohemia, Branišovská, České Budějovice, Czech Republic
| | | | - Rafael Prados-Rosales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Estíbaliz Atondo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Itziar Martín
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Aize Pellón
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Félix Elortza
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
- * E-mail:
| |
Collapse
|
22
|
de Marcken M, Dhaliwal K, Danielsen AC, Gautron AS, Dominguez-Villar M. TLR7 and TLR8 activate distinct pathways in monocytes during RNA virus infection. Sci Signal 2019; 12:12/605/eaaw1347. [DOI: 10.1126/scisignal.aaw1347] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human blood CD14+monocytes are bone marrow–derived white blood cells that sense and respond to pathogens. Although innate immune activation by RNA viruses preferentially occurs through intracellular RIG-I–like receptors, other nucleic acid recognition receptors, such as Toll-like receptors (TLRs), play a role in finely programming the final outcome of virus infection. Here, we dissected how human monocytes respond to infection with either Coxsackie (CV), encephalomyocarditis (EMCV), influenza A (IAV), measles (MV), Sendai (SV), or vesicular stomatitis (VSV) virus. We found that in monocytes, type I interferon (IFN) and cytokine responses to infection were RNA virus specific and differentially involved TLR7 and TLR8, which sense single-stranded RNA. These TLRs activated distinct signaling cascades in monocytes, which correlated with differences in the production of cytokines involved in the polarization of CD4+T helper cells. Furthermore, we found that TLR7 signaling specifically increased expression of the transcription factor FOSL1, which reduced IL-27 and TNFα production by monocytes. TLR7, but not TLR8, activation of monocytes also stimulated Ca2+flux that prevented type I IFN responses. Our work demonstrates that in human monocytes, TLR7 and TLR8 triggered different signaling pathways that contribute to distinct phenotypes during RNA virus infection. In addition, we defined individual targets within these pathways that promoted specific T helper and antiviral responses.
Collapse
Affiliation(s)
- Marine de Marcken
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Khushwant Dhaliwal
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
23
|
Cytokine Expression Patterns and Single Nucleotide Polymorphisms (SNPs) in Patients with Chronic Borreliosis. Antibiotics (Basel) 2019; 8:antibiotics8030107. [PMID: 31366164 PMCID: PMC6784230 DOI: 10.3390/antibiotics8030107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/21/2019] [Accepted: 07/25/2019] [Indexed: 12/19/2022] Open
Abstract
(1) Background: Genetically based hyperinflammation may play a role in pathogen defense. We here questioned whether alterations in circulating monocytes/macrophages, inflammatory biomarkers and a functional SNP (single nucleotide polymorphisms) of the Interleukin-6 (IL-6) promotor might play a role in patients with persistent, and treatment resistant borreliosis. (2) Methods: Leukocyte subpopulations were studied by flow cytometry; plasma cytokines were determined by a chemiluminescence based ELISA (Immulite®), and genotypes of the IL-6 promotor SNP rs1800795 were determined by pyrosequencing. (3) Results: In a cohort of n = 107 Lyme borreliosis patients, who concomitantly manifested either malignant diseases (group 1), autoimmune disorders (group 2), neurological diseases (group 3), or morbidities caused by multiple other infectious complications (group 4), we found decreased numbers of anti-inflammatory CD163-positive macrophages, elevated concentrations of inflammatory cytokines, and an imbalance of IL-6 promotor SNP rs1800795 genotypes. The most prominently upregulated cytokines were IL-1β, and IL-8. (4) Conclusions: Increased pro-inflammatory phenotypes identified by monocyte/macrophage subtypes and concomitantly increased cytokines appear to be valid to monitor disease activity in patients with persistent Lyme borreliosis. Patterns may vary by additional co-morbidities. In patients with autoimmune diseases, increased frequencies of a heterozygous IL-6 promotor SNP rs1800795 were identified. This functional SNP may guide chronic inflammation, impacting other cytokines to trigger trigger chronicity and therapeutic resistance in Lyme borreliosis.
Collapse
|
24
|
Cervantes JL, Oak E, Garcia J, Liu H, Lorenzini PA, Batra D, Chhabra A, Salazar JC, Roca X. Vitamin D modulates human macrophage response to Mycobacterium tuberculosis DNA. Tuberculosis (Edinb) 2019; 116S:S131-S137. [PMID: 31085128 PMCID: PMC6626683 DOI: 10.1016/j.tube.2019.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is a facultative intracellular pathogen that infects macrophages where it avoids elimination by interfering with host defense mechanisms, including phago-lysosome fusion. Endosomal Toll-like receptors (TLRs) generate Type I Interferons (IFNs), which are associated with active tuberculosis (TB). We aimed to explore if DNA from different Mtb lineages lead to differences in the inflammatory response of human monocytic/macrophage cells. THP-1 cells which express two inducible reporter constructs for interferons (IFNs) as well as for NF-κB, were stimulated via endosomal delivery of Mtb DNA as a nanocomplex with PEI. DNA from different Mtb phylogenetic lineages elicited differential inflammatory responses in human macrophages. An initial relatively weak IRF-mediated response to DNA from HN878 and H37Rv increased if the cells were pre-treated with Vitamin D (Vit D) for 72 h. RNAseq of THP-1 under different transformation conditions showed that pre-treatment with Vit D upregulated several TLR9 variants, as well as genes involved in inflammatory immune response to infection, immune cell activation, Type I IFN regulation, and regulation of inflammation. Vit D appears to be important in increasing low IRF responses to DNA from certain lineages of Mtb. Variations in the IRF-mediated response to DNA derived from different Mtb genotypes are potentially important in the pathogenesis of tuberculosis since Type I IFN responses are associated with active disease. The role of Vit D in these responses could also translate into future therapeutic approaches.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX, USA.
| | - Esther Oak
- University of New England, College of Dental Medicine, Portland, ME, USA
| | - John Garcia
- University of Connecticut Health, School of Public Health, Farmington, CT, USA
| | - Hongfei Liu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Paolo A Lorenzini
- School of Biological Sciences, Nanyang Technological University, Singapore; Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate School, Nanyang Technological University, Singapore
| | - Deepika Batra
- Stem Cell Institute, Amity University Haryana, Manesar, Gurugram, Haryana, India
| | - Arvind Chhabra
- Stem Cell Institute, Amity University Haryana, Manesar, Gurugram, Haryana, India
| | - Juan C Salazar
- University of Connecticut Health, Department of Pediatrics, Farmington, CT, USA; Connecticut Children's Medical Center, Hartford, CT, USA
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
25
|
Vierbuchen T, Stein K, Heine H. RNA is taking its Toll: Impact of RNA-specific Toll-like receptors on health and disease. Allergy 2019; 74:223-235. [PMID: 30475385 DOI: 10.1111/all.13680] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/08/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
RNA-sensing Toll-like receptors (TLRs) are often described as antiviral receptors of the innate immune system. However, the past decade has shown that the function and relevance of these receptors are far more complex. They were found to be essential for the detection of various bacterial, archaeal, and eukaryotic microorganisms and facilitate the discrimination between dead and living microbes. The cytokine and interferon response profile that is triggered has the potential to improve the efficacy of next-generation vaccines and may prevent the development of asthma and allergy. Nevertheless, the ability to recognize foreign RNA comes with a cost as also damaged host cells can release nucleic acids that might induce an inappropriate immune response. Thus, it is not surprising that RNA-sensing TLRs play a key role in various autoimmune diseases. However, promising new inhibitors and antagonists are on the horizon to improve their treatment.
Collapse
Affiliation(s)
- Tim Vierbuchen
- Division of Innate Immunity Research Center Borstel – Leibniz Lung Center Borstel Germany
| | - Karina Stein
- Division of Innate Immunity Research Center Borstel – Leibniz Lung Center Borstel Germany
- Airway Research Center North (ARCN) German Center for Lung Research (DZL) Borstel Germany
| | - Holger Heine
- Division of Innate Immunity Research Center Borstel – Leibniz Lung Center Borstel Germany
- Airway Research Center North (ARCN) German Center for Lung Research (DZL) Borstel Germany
| |
Collapse
|
26
|
Heil M, Vega-Muñoz I. Nucleic Acid Sensing in Mammals and Plants: Facts and Caveats. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 345:225-285. [PMID: 30904194 DOI: 10.1016/bs.ircmb.2018.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The accumulation of nucleic acids in aberrant compartments is a signal of danger: fragments of cytosolic or extracellular self-DNA indicate cellular dysfunctions or disruption, whereas cytosolic fragments of nonself-DNA or RNA indicate infections. Therefore, nucleic acids trigger immunity in mammals and plants. In mammals, endosomal Toll-like receptors (TLRs) sense single-stranded (ss) or double-stranded (ds) RNA or CpG-rich DNA, whereas various cytosolic receptors sense dsDNA. Although a self/nonself discrimination could favor targeted immune responses, no sequence-specific sensing of nucleic acids has been reported for mammals. Specific immune responses to extracellular self-DNA versus DNA from related species were recently reported for plants, but the underlying mechanism remains unknown. The subcellular localization of mammalian receptors can favor self/nonself discrimination based on the localization of DNA fragments. However, autoantibodies and diverse damage-associated molecular patterns (DAMPs) shuttle DNA through membranes, and most of the mammalian receptors share downstream signaling elements such as stimulator of interferon genes (STING) and the master transcription regulators, nuclear factor (NF)-κB, and interferon regulatory factor 3 (IRF3). The resulting type I interferon (IFN) response stimulates innate immunity against multiple threats-from infection to physical injury or endogenous DNA damage-all of which lead to the accumulation of eDNA or cytoplasmatic dsDNA. Therefore, no or only low selective pressures might have favored a strict self/nonself discrimination in nucleic acid sensing. We conclude that the discrimination between self- and nonself-DNA is likely to be less strict-and less important-than assumed originally.
Collapse
Affiliation(s)
- Martin Heil
- Departmento de Ingeniería Genética, CINVESTAV-Irapuato, Irapuato, Guanajuato, Mexico.
| | - Isaac Vega-Muñoz
- Departmento de Ingeniería Genética, CINVESTAV-Irapuato, Irapuato, Guanajuato, Mexico
| |
Collapse
|
27
|
Tamtaji OR, Mobini M, Reiter RJ, Azami A, Gholami MS, Asemi Z. Melatonin, a toll-like receptor inhibitor: Current status and future perspectives. J Cell Physiol 2018; 234:7788-7795. [PMID: 30387141 DOI: 10.1002/jcp.27698] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are crucial activators of inflammatory responses, they are considered immune receptors. TLRs are of fundamental importance in the pathophysiology of disorders related to inflammation including neurodegenerative diseases and cancer. Melatonin is a beneficial agent in the treatment of inflammatory and immune disorders. Melatonin is potent anti-inflammatory hormone that regulates various molecular pathways. Withal, limited studies have evaluated the inhibitory role of melatonin on TLRs. This review summarizes the current knowledge related to the effects of melatonin on TLRs in some common inflammatory and immunity disorders.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Halal Research Center of IRI, FDA, Tehran, Iran.,Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Mobini
- Kinesiology Department, University of Calgary, Calgary, Alberta, Canada
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Abolfazl Azami
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Saeed Gholami
- Skull Base Research Center, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Student Research Committee, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
28
|
Abstract
Pattern recognition receptors (PRRs) survey intra- and extracellular spaces for pathogen-associated molecular patterns (PAMPs) within microbial products of infection. Recognition and binding to cognate PAMP ligand by specific PRRs initiates signaling cascades that culminate in a coordinated intracellular innate immune response designed to control infection. In particular, our immune system has evolved specialized PRRs to discriminate viral nucleic acid from host. These are critical sensors of viral RNA to trigger innate immunity in the vertebrate host. Different families of PRRs of virus infection have been defined and reveal a diversity of PAMP specificity for wide viral pathogen coverage to recognize and extinguish virus infection. In this review, we discuss recent insights in pathogen recognition by the RIG-I-like receptors, related RNA helicases, Toll-like receptors, and other RNA sensor PRRs, to present emerging themes in innate immune signaling during virus infection.
Collapse
Affiliation(s)
- Kwan T Chow
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| | - Michael Gale
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| | - Yueh-Ming Loo
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| |
Collapse
|
29
|
A multi-omic analysis reveals the regulatory role of CD180 during the response of macrophages to Borrelia burgdorferi. Emerg Microbes Infect 2018; 7:19. [PMID: 29511161 PMCID: PMC5841238 DOI: 10.1038/s41426-017-0018-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Macrophages are cells of the innate immune system with the ability to phagocytose and induce a global pattern of responses that depend on several signaling pathways. We have determined the biosignature of murine bone marrow-derived macrophages and human blood monocytes using transcriptomic and proteomic approaches. We identified a common pattern of genes that are transcriptionally regulated and overall indicate that the response to B. burgdorferi involves the interaction of spirochetal antigens with several inflammatory pathways corresponding to primary (triggered by pattern-recognition receptors) and secondary (induced by proinflammatory cytokines) responses. We also show that the Toll-like receptor family member CD180 is downregulated by the stimulation of macrophages, but not monocytes, with the spirochete. Silencing Cd180 results in increased phagocytosis while tempering the production of the proinflammatory cytokine TNF. Cd180-silenced cells produce increased levels of Itgam and surface CD11b, suggesting that the regulation of CD180 by the spirochete initiates a cascade that increases CR3-mediated phagocytosis of the bacterium while repressing the consequent inflammatory response.
Collapse
|
30
|
Paquette JK, Ma Y, Fisher C, Li J, Lee SB, Zachary JF, Kim YS, Teuscher C, Weis JJ. Genetic Control of Lyme Arthritis by Borrelia burgdorferi Arthritis-Associated Locus 1 Is Dependent on Localized Differential Production of IFN-β and Requires Upregulation of Myostatin. THE JOURNAL OF IMMUNOLOGY 2017; 199:3525-3534. [PMID: 28986440 DOI: 10.4049/jimmunol.1701011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/14/2017] [Indexed: 01/29/2023]
Abstract
Previously, using a forward genetic approach, we identified differential expression of type I IFN as a positional candidate for an expression quantitative trait locus underlying Borrelia burgdorferi arthritis-associated locus 1 (Bbaa1). In this study, we show that mAb blockade revealed a unique role for IFN-β in Lyme arthritis development in B6.C3-Bbaa1 mice. Genetic control of IFN-β expression was also identified in bone marrow-derived macrophages stimulated with B. burgdorferi, and it was responsible for feed-forward amplification of IFN-stimulated genes. Reciprocal radiation chimeras between B6.C3-Bbaa1 and C57BL/6 mice revealed that arthritis is initiated by radiation-sensitive cells, but orchestrated by radiation-resistant components of joint tissue. Advanced congenic lines were developed to reduce the physical size of the Bbaa1 interval, and confirmed the contribution of type I IFN genes to Lyme arthritis. RNA sequencing of resident CD45- joint cells from advanced interval-specific recombinant congenic lines identified myostatin as uniquely upregulated in association with Bbaa1 arthritis development, and myostatin expression was linked to IFN-β production. Inhibition of myostatin in vivo suppressed Lyme arthritis in the reduced interval Bbaa1 congenic mice, formally implicating myostatin as a novel downstream mediator of the joint-specific inflammatory response to B. burgdorferi.
Collapse
Affiliation(s)
- Jackie K Paquette
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Ying Ma
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Colleen Fisher
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Jinze Li
- Department of Pathology, University of Utah, Salt Lake City, UT 84112
| | - Sang Beum Lee
- Department of Human Nutrition, Food, and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI 96822
| | - James F Zachary
- Department of Veterinary Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61820; and
| | - Yong Soo Kim
- Department of Human Nutrition, Food, and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI 96822
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Janis J Weis
- Department of Pathology, University of Utah, Salt Lake City, UT 84112;
| |
Collapse
|
31
|
Hawley KL, Cruz AR, Benjamin SJ, La Vake CJ, Cervantes JL, LeDoyt M, Ramirez LG, Mandich D, Fiel-Gan M, Caimano MJ, Radolf JD, Salazar JC. IFNγ Enhances CD64-Potentiated Phagocytosis of Treponema pallidum Opsonized with Human Syphilitic Serum by Human Macrophages. Front Immunol 2017; 8:1227. [PMID: 29051759 PMCID: PMC5633599 DOI: 10.3389/fimmu.2017.01227] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022] Open
Abstract
Syphilis is a multi-stage, sexually transmitted disease caused by the spirochete Treponema pallidum (Tp). Considered broadly, syphilis can be conceptualized as a dualistic process in which spirochete-driven inflammation, the cause of clinical manifestations, coexists to varying extents with bacterial persistence. Inflammation is elicited in the tissues, along with the persistence of spirochetes to keep driving a robust immune response while evading host defenses; this duality is best exemplified during the florid, disseminated stage called secondary syphilis (SS). SS lesions typically contain copious amounts of spirochetes along with a mixed cellular infiltrate consisting of CD4+ T cells, CD8+ T cells, NK cells, plasma cells, and macrophages. In the rabbit model, Tp are cleared by macrophages via antibody-mediated opsonophagocytosis. Previously, we demonstrated that human syphilitic serum (HSS) promotes efficient uptake of Tp by human monocytes and that opsonophagocytosis of Tp markedly enhances cytokine production. Herein, we used monocyte-derived macrophages to study Tp–macrophage interactions ex vivo. In the absence of HSS, monocyte-derived macrophages internalized low numbers of Tp and secreted little cytokine (e.g., TNF). By contrast, these same macrophages internalized large numbers of unopsonized Borrelia burgdorferi and secreted robust levels of cytokines. Maturation of macrophages with M-CSF and IFNγ resulted in a macrophage phenotype with increased expression of HLA-DR, CD14, inducible nitric oxide synthase, TLR2, TLR8, and the Fcγ receptors (FcγR) CD64 and CD16, even in the absence of LPS. Importantly, IFNγ-polarized macrophages resulted in a statistically significant increase in opsonophagocytosis of Tp accompanied by enhanced production of cytokines, macrophage activation markers (CD40, CD80), TLRs (TLR2, TLR7, TLR8), chemokines (CCL19, CXCL10, CXCL11), and TH1-promoting cytokines (IL-12, IL-15). Finally, the blockade of FcγRs, primarily CD64, significantly diminished spirochetal uptake and proinflammatory cytokine secretion by IFNγ-stimulated macrophages. Our ex vivo studies demonstrate the importance of CD64-potentiated uptake of opsonized Tp and suggest that IFNγ-activated macrophages have an important role in the context of early syphilis. Our study results also provide an ex vivo surrogate system for use in future syphilis vaccine studies.
Collapse
Affiliation(s)
- Kelly L Hawley
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States
| | - Adriana R Cruz
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Sarah J Benjamin
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Department of Immunology, UConn Health, Farmington, CT, United States
| | - Carson J La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Jorge L Cervantes
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States
| | - Morgan LeDoyt
- Department of Medicine, UConn Health, Farmington, CT, United States
| | - Lady G Ramirez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Daniza Mandich
- Department of Pathology, Hartford Hospital, Hartford, CT, United States
| | - Mary Fiel-Gan
- Department of Pathology, Hartford Hospital, Hartford, CT, United States
| | | | - Justin D Radolf
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia.,Department of Immunology, UConn Health, Farmington, CT, United States.,Department of Medicine, UConn Health, Farmington, CT, United States.,Department of Genetics and Developmental Biology, UConn Health, Farmington, CT, United States
| | - Juan C Salazar
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia.,Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
32
|
Phagocytic Receptors Activate Syk and Src Signaling during Borrelia burgdorferi Phagocytosis. Infect Immun 2017; 85:IAI.00004-17. [PMID: 28717031 DOI: 10.1128/iai.00004-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022] Open
Abstract
Phagocytosis of the Lyme disease-causing pathogen Borrelia burgdorferi has been shown to be important for generating an inflammatory response to the pathogen. As a result, understanding the mechanisms of phagocytosis has been an area of great interest in the field of Lyme disease. Several cell surface receptors that participate in B. burgdorferi phagocytosis have been reported, including the scavenger receptor MARCO and integrin α3β1. We sought to define the mechanisms by which these receptors mediate phagocytosis and to identify signaling pathways activated downstream of these receptors upon contact with B. burgdorferi We identified both Syk and Src signaling pathways as ones that participate in B. burgdorferi phagocytosis and the resulting cytokine activation. In our studies, we found that both MARCO and integrin β1 play a role in the activation of the Src kinase pathway. However, only integrin β1 participates in the activation of Syk. Interestingly, the integrin activates Syk without the help of the signaling adaptor Dap12 or FcRγ. Thus, we report that multiple pathways participate in B. burgdorferi internalization and that different cell surface receptors act simultaneously in cooperation and independently to mediate phagocytosis.
Collapse
|
33
|
Aslam B, Nisar MA, Khurshid M, Farooq Salamat MK. Immune escape strategies of Borrelia burgdorferi. Future Microbiol 2017; 12:1219-1237. [PMID: 28972415 DOI: 10.2217/fmb-2017-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The borrelial resurge demonstrates that Borrelia burgdorferi is a persistent health problem. This spirochete is responsible for a global public health concern called Lyme disease. B. burgdorferi faces diverse environmental conditions of its vector and host during its life cycle. To circumvent the host immune system is a prominent feature of B. burgdorferi. To date, numerous studies have reported on the various mechanisms used by this pathogen to evade the host defense mechanisms. This current review attempts to consolidate this information to describe the immunological and molecular methods used by B. burgdorferi for its survival.
Collapse
Affiliation(s)
- Bilal Aslam
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Atif Nisar
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Mohsin Khurshid
- Department of Microbiology, Government College University, Faisalabad, Pakistan.,College of Allied Health Professionals, Directorate of Medical Sciences, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
34
|
Cervantes J. Doctor says you are cured, but you still feel the pain. Borrelia DNA persistence in Lyme disease. Microbes Infect 2017. [DOI: 10.1016/j.micinf.2017.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
35
|
Gidon A, Åsberg SE, Louet C, Ryan L, Haug M, Flo TH. Persistent mycobacteria evade an antibacterial program mediated by phagolysosomal TLR7/8/MyD88 in human primary macrophages. PLoS Pathog 2017; 13:e1006551. [PMID: 28806745 PMCID: PMC5570494 DOI: 10.1371/journal.ppat.1006551] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/24/2017] [Accepted: 07/25/2017] [Indexed: 12/20/2022] Open
Abstract
Pathogenic mycobacteria reside in macrophages where they avoid lysosomal targeting and degradation through poorly understood mechanisms proposed to involve arrest of phagosomal maturation at an early endosomal stage. A clear understanding of how this relates to host defenses elicited from various intracellular compartments is also missing and can only be studied using techniques allowing single cell and subcellular analyses. Using confocal imaging of human primary macrophages infected with Mycobacterium avium (Mav) we show evidence that Mav phagosomes are not arrested at an early endosomal stage, but mature to a (LAMP1+/LAMP2+/CD63+) late endosomal/phagolysosomal stage where inflammatory signaling and Mav growth restriction is initiated through a mechanism involving Toll-like receptors (TLR) 7 and 8, the adaptor MyD88 and transcription factors NF-κB and IRF-1. Furthermore, a fraction of the mycobacteria re-establish in a less hostile compartment (LAMP1-/LAMP2-/CD63-) where they not only evade destruction, but also recognition by TLRs, growth restriction and inflammatory host responses that could be detrimental for intracellular survival and establishment of chronic infections. Mycobacterium avium is increasingly reported as a causative agent of non-tuberculous disease in immunocompromised patients and in individuals with underlying disease or using immunosuppressant drugs, with prevalence often higher than the more pathogenic M. tuberculosis in developed countries. Both M. avium and M. tuberculosis cause persistent infections by surviving inside host macrophages. Here, we identify from which compartment M. avium evoke inflammatory signaling in human primary macrophages, and the pattern-recognition receptors involved. In essence, we present three key findings: 1) M. avium phagosomes are not arrested at an early endosomal stage, but rather mature normally into phagolysosomes from where a fraction of the bacteria escape and re-establish in a new compartment. 2) In addition to avoiding degradation in phagolysosomes, by escaping M. avium also evade inflammatory signaling. 3) M. avium unable to escape is degraded in phagolysosomes and recognized by Toll-like receptors 7 and 8. Our results can contribute to new understanding of intracellular infections, and thus have vital clinical implications for development of novel anti-microbial strategies and host-targeted therapy to mycobacterial and other infectious diseases.
Collapse
Affiliation(s)
- Alexandre Gidon
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Signe Elisabeth Åsberg
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Claire Louet
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Liv Ryan
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Markus Haug
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- The Central Norway Regional Health Authority, Trondheim, Norway
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail:
| |
Collapse
|
36
|
Kieser KJ, Kagan JC. Multi-receptor detection of individual bacterial products by the innate immune system. Nat Rev Immunol 2017; 17:376-390. [PMID: 28461704 DOI: 10.1038/nri.2017.25] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The receptors of the innate immune system detect specific microbial ligands to promote effective inflammatory and adaptive immune responses. Although this idea is well appreciated, studies in recent years have highlighted the complexity of innate immune detection, with multiple host receptors recognizing the same microbial ligand. Understanding the collective actions of diverse receptors that recognize common microbial signatures represents a new frontier in the study of innate immunity, and is the focus of this Review. Here, we discuss examples of individual bacterial cell wall components that are recognized by at least two and as many as four different receptors of the innate immune system. These receptors survey the extracellular or cytosolic spaces for their cognate ligands and operate in a complementary manner to induce distinct cellular responses. We further highlight that, despite this genetic diversity in receptors and pathways, common features exist to explain the operation of these receptors. These common features may help to provide unifying organizing principles associated with host defence.
Collapse
Affiliation(s)
- Karen J Kieser
- Department of Pediatrics, Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Jonathan C Kagan
- Department of Pediatrics, Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
37
|
Abstract
Lipoproteins are lipid-modified proteins that dominate the spirochetal proteome. While found in all bacteria, spirochetal lipoproteins have unique features and play critical roles in spirochete biology. For this reason, considerable effort has been devoted to determining how the lipoproteome is generated. Essential features of the structural elements of lipoproteins are now understood with greater clarity, enabling greater confidence in identification of lipoproteins from genomic sequences. The journey from the ribosome to the outer membrane, and in some cases, to the cellular surface has been defined, including secretion, lipidation, sorting, and export across the outer membrane. Given their abundance and importance, it is not surprising that spirochetes have developed a number of strategies for regulating the spatiotemporal expression of lipoproteins. In some cases, lipoprotein expression is tied to various environmental cues, while in other cases, it is linked to growth rate. This regulation enables spirochetes to express certain lipoproteins at high levels in one phase of the spirochete lifecycle, while dramatically downregulating the same lipoproteins in other phases. The mammalian host has developed specialized mechanisms for recognizing lipoproteins and triggering an immune response. Evasion of that immune response is essential for spirochete persistence. For this reason, spirochetes have developed mechanisms for altering lipoproteins. Lipoproteins recognized by antibodies formed during infection are key serodiagnostic antigens. In addition, lipoprotein vaccines have been developed for generating an immune response to control or prevent a spirochete infection. This chapter summarizes our current understanding of lipoproteins in interactions of spirochetes with their hosts.
Collapse
|
38
|
Tan Y, Kagan JC. Microbe-inducible trafficking pathways that control Toll-like receptor signaling. Traffic 2017; 18:6-17. [PMID: 27731905 PMCID: PMC5182131 DOI: 10.1111/tra.12454] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
The receptors of the mammalian innate immune system are designed for rapid microbial detection, and are located in organelles that are conducive to serve these needs. However, emerging evidence indicates that the sites of microbial detection are not the sites of innate immune signal transduction. Rather, microbial detection triggers the movement of receptors to regions of the cell where factors called sorting adaptors detect active receptors and promote downstream inflammatory responses. These findings highlight the critical role that membrane trafficking pathways play in the initiation of innate immunity to infection. In this review, we describe pathways that promote the microbe-inducible endocytosis of Toll-like receptors (TLRs), and the microbe-inducible movement of TLRs between intracellular compartments. We highlight a new class of proteins called Transporters Associated with the eXecution of Inflammation (TAXI), which have the unique ability to transport TLRs and their microbial ligands to signaling-competent regions of the cell, and we discuss the means by which the subcellular sites of signal transduction are defined.
Collapse
Affiliation(s)
- Yunhao Tan
- Harvard Medical School and Division of Gastroenterology, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Jonathan C. Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children’s Hospital, Boston, MA, 02115, USA
| |
Collapse
|
39
|
Abstract
Lyme borreliosis is a tick-borne disease that predominantly occurs in temperate regions of the northern hemisphere and is primarily caused by the bacterium Borrelia burgdorferi in North America and Borrelia afzelii or Borrelia garinii in Europe and Asia. Infection usually begins with an expanding skin lesion, known as erythema migrans (referred to as stage 1), which, if untreated, can be followed by early disseminated infection, particularly neurological abnormalities (stage 2), and by late infection, especially arthritis in North America or acrodermatitis chronica atrophicans in Europe (stage 3). However, the disease can present with any of these manifestations. During infection, the bacteria migrate through the host tissues, adhere to certain cells and can evade immune clearance. Yet, these organisms are eventually killed by both innate and adaptive immune responses and most inflammatory manifestations of the infection resolve. Except for patients with erythema migrans, Lyme borreliosis is diagnosed based on a characteristic clinical constellation of signs and symptoms with serological confirmation of infection. All manifestations of the infection can usually be treated with appropriate antibiotic regimens, but the disease can be followed by post-infectious sequelae in some patients. Prevention of Lyme borreliosis primarily involves the avoidance of tick bites by personal protective measures.
Collapse
Affiliation(s)
- Allen C Steere
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, USA
- Harvard Medical School, Harvard University, Boston, Massachusetts, USA
| | - Franc Strle
- Department of Infectious Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Gary P Wormser
- Division of Infectious Diseases, New York Medical College, Valhalla, New York, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - John A Branda
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joppe W R Hovius
- Center for Experimental and Molecular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Xin Li
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, Massachusetts, USA
| | - Paul S Mead
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| |
Collapse
|
40
|
Kurioka A, Walker LJ, Klenerman P, Willberg CB. MAIT cells: new guardians of the liver. Clin Transl Immunology 2016; 5:e98. [PMID: 27588203 PMCID: PMC5007630 DOI: 10.1038/cti.2016.51] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 02/08/2023] Open
Abstract
The liver is an important immunological organ that remains sterile and tolerogenic in homeostasis, despite continual exposure to non-self food and microbial-derived products from the gut. However, where intestinal mucosal defenses are breached or in the presence of a systemic infection, the liver acts as a second 'firewall', because of its enrichment with innate effector cells able to rapidly respond to infections or tissue dysregulation. One of the largest populations of T cells within the human liver are mucosal-associated invariant T (MAIT) cells, a novel innate-like T-cell population that can recognize a highly conserved antigen derived from the microbial riboflavin synthesis pathway. MAIT cells are emerging as significant players in the human immune system, associated with an increasing number of clinical diseases of bacterial, viral, autoimmune and cancerous origin. As reviewed here, we are only beginning to investigate the potential role of this dominant T-cell subset in the liver, but the reactivity of MAIT cells to both inflammatory cytokines and riboflavin derivatives suggests that MAIT cells may have an important role in first line of defense as part of the liver firewall. As such, MAIT cells are promising targets for modulating the host defense and inflammation in both acute and chronic liver diseases.
Collapse
Affiliation(s)
- Ayako Kurioka
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Lucy J Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Christian B Willberg
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Stein K, Brand S, Jenckel A, Sigmund A, Chen ZJ, Kirschning CJ, Kauth M, Heine H. Endosomal recognition of Lactococcus lactis G121 and its RNA by dendritic cells is key to its allergy-protective effects. J Allergy Clin Immunol 2016; 139:667-678.e5. [PMID: 27544739 DOI: 10.1016/j.jaci.2016.06.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 06/02/2016] [Accepted: 06/13/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Bacterial cowshed isolates are allergy protective in mice; however, the underlying mechanisms are largely unknown. We examined the ability of Lactococcus lactis G121 to prevent allergic inflammatory reactions. OBJECTIVE We sought to identify the ligands and pattern recognition receptors through which L lactis G121 confers allergy protection. METHODS L lactis G121-induced cytokine release and surface expression of costimulatory molecules by untreated or inhibitor-treated (bafilomycin and cytochalasin D) human monocyte-derived dendritic cells (moDCs), bone marrow-derived mouse dendritic cells (BMDCs), and moDC/naive CD4+ T-cell cocultures were analyzed by using ELISA and flow cytometry. The pathology of ovalbumin-induced acute allergic airway inflammation after adoptive transfer of BMDCs was examined by means of microscopy. RESULTS L lactis G121-treated murine BMDCs and human moDCs released TH1-polarizing cytokines and induced TH1 T cells. Inhibiting phagocytosis and endosomal acidification in BMDCs or moDCs impaired the release of TH1-polarizing cytokines, costimulatory molecule expression, and T-cell activation on L lactis G121 challenge. In vivo allergy protection mediated by L lactis G121 was dependent on endosomal acidification in dendritic cells (DCs). Toll-like receptor (Tlr) 13-/- BMDCs showed a weak response to L lactis G121 and were unresponsive to its RNA. The TH1-polarizing activity of L lactis G121-treated human DCs was blocked by TLR8-specific inhibitors, mediated by L lactis G121 RNA, and synergistically enhanced by activation of nucleotide-binding oligomerization domain-containing protein (NOD) 2. CONCLUSION Bacterial RNA is the main driver of L lactis G121-mediated protection against experimentally induced allergy and requires both bacterial uptake by DCs and endosomal acidification. In mice L lactis G121 RNA signals through TLR13; however, the most likely intracellular receptor in human subjects is TLR8.
Collapse
Affiliation(s)
- Karina Stein
- Division of Innate Immunity, Research Center Borstel, Airway Research Center North, German Center for Lung Research (DZL), Germany
| | | | - André Jenckel
- Division of Innate Immunity, Research Center Borstel, Airway Research Center North, German Center for Lung Research (DZL), Germany
| | - Anna Sigmund
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Zhijian James Chen
- Department of Molecular Biology, Howard Hughes Medical Institute, UT Southwestern Medical School, Dallas, Tex
| | | | | | - Holger Heine
- Division of Innate Immunity, Research Center Borstel, Airway Research Center North, German Center for Lung Research (DZL), Germany.
| |
Collapse
|
42
|
Scholl DC, Embers ME, Caskey JR, Kaushal D, Mather TN, Buck WR, Morici LA, Philipp MT. Immunomodulatory effects of tick saliva on dermal cells exposed to Borrelia burgdorferi, the agent of Lyme disease. Parasit Vectors 2016; 9:394. [PMID: 27391120 PMCID: PMC4938952 DOI: 10.1186/s13071-016-1638-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/10/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The prolonged feeding process of ixodid ticks, in combination with bacterial transmission, should lead to a robust inflammatory response at the blood-feeding site. Yet, factors present in tick saliva may down-regulate such responses, which may be beneficial to spirochete transmission. The primary goal of this study was to test the hypothesis that tick saliva, in the context of Borrelia burgdorferi, can have widespread effects on the production of immune mediators in skin. METHODS A cross-section of tick feeding on skin was examined histologically. Human THP-1 cells stimulated with B. burgdorferi and grown in the presence or absence of tick saliva were examined by human DNA microarray, cytokine bead array, sandwich ELISA, and qRT-PCR. Similar experiments were also conducted using dermal fibroblasts. RESULTS Tick feeding on skin showed dermal infiltration of histiocytes and granulocytes at the bite location. Changes in monocytic transcript levels during co-culture with B. burgdorferi and saliva indicated that tick saliva had a suppressive effect on the expression of certain pro-inflammatory mediators, such as IL-8 (CXCL8) and TLR2, but had a stimulatory effect on specific molecules such as the Interleukin 10 receptor, alpha subunit (IL-10RA), a known mediator of the immunosuppressive signal of IL-10. Stimulated cell culture supernatants were analyzed via antigen-capture ELISA and cytokine bead array for inflammatory mediator production. Treatment of monocytes with saliva significantly reduced the expression of several key mediators including IL-6, IL-8 and TNF-alpha. Tick saliva had an opposite effect on dermal fibroblasts. Rather than inhibiting, saliva enhanced production of pro-inflammatory mediators, including IL-8 and IL-6 from these sentinel skin cells. CONCLUSIONS The effects of ixodid tick saliva on resident skin cells is cell type-dependent. The response to both tick and pathogen at the site of feeding favors pathogen transmission, but may not be wholly suppressed by tick saliva.
Collapse
Affiliation(s)
- Dorothy C. Scholl
- />Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA USA
- />Present Address: Department of Biology, University of New Mexico, Albuquerque, NM USA
| | - Monica E. Embers
- />Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA USA
| | - John R. Caskey
- />Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA USA
| | - Deepak Kaushal
- />Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA USA
| | - Thomas N. Mather
- />Center for Vector-Borne Disease, University of Rhode Island, Kingston, RI USA
| | - Wayne R. Buck
- />Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana USA
- />Present Address: AbbVie, 1 N Waukegan Rd, North Chicago, IL USA
| | - Lisa A. Morici
- />Department of Microbiology and Immunology, Tulane University Medical School, New Orleans, LA USA
| | - Mario T. Philipp
- />Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA USA
| |
Collapse
|
43
|
Rahman S, Shering M, Ogden NH, Lindsay R, Badawi A. Toll-like receptor cascade and gene polymorphism in host-pathogen interaction in Lyme disease. J Inflamm Res 2016; 9:91-102. [PMID: 27330321 PMCID: PMC4898433 DOI: 10.2147/jir.s104790] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lyme disease (LD) risk occurs in North America and Europe where the tick vectors of the causal agent Borrelia burgdorferi sensu lato are found. It is associated with local and systemic manifestations, and has persistent posttreatment health complications in some individuals. The innate immune system likely plays a critical role in both host defense against B. burgdorferi and disease severity. Recognition of B. burgdorferi, activation of the innate immune system, production of proinflammatory cytokines, and modulation of the host adaptive responses are all initiated by Toll-like receptors (TLRs). A number of Borrelia outer-surface proteins (eg, OspA and OspB) are recognized by TLRs. Specifically, TLR1 and TLR2 were identified as the receptors most relevant to LD. Several functional single-nucleotide polymorphisms have been identified in TLR genes, and are associated with varying cytokines types and synthesis levels, altered pathogen recognition, and disruption of the downstream signaling cascade. These single-nucleotide polymorphism-related functional alterations are postulated to be linked to disease development and posttreatment persistent illness. Elucidating the role of TLRs in LD may facilitate a better understanding of disease pathogenesis and can provide an insight into novel therapeutic targets during active disease or postinfection and posttreatment stages.
Collapse
Affiliation(s)
- Shusmita Rahman
- National Microbiology Laboratory, Public Health Agency of Canada, Toronto, ON, Canada
| | - Maria Shering
- Faculty of Arts and Science, University of Toronto, Toronto, ON, Canada
| | - Nicholas H Ogden
- National Microbiology Laboratory, Public Health Agency of Canada, Saint-Hyacinthe, QC, Canada
| | - Robbin Lindsay
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Alaa Badawi
- National Microbiology Laboratory, Public Health Agency of Canada, Toronto, ON, Canada
| |
Collapse
|
44
|
Toll-like receptors and chronic inflammation in rheumatic diseases: new developments. Nat Rev Rheumatol 2016; 12:344-57. [PMID: 27170508 DOI: 10.1038/nrrheum.2016.61] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In the past few years, new developments have been reported on the role of Toll-like receptors (TLRs) in chronic inflammation in rheumatic diseases. The inhibitory function of TLR10 has been demonstrated. Receptors that enhance the function of TLRs, and several TLR inhibitors, have been identified. In addition, the role of the microbiome and TLRs in the onset of rheumatic diseases has been reported. We review novel insights on the role of TLRs in several inflammatory joint diseases, including rheumatoid arthritis, systemic lupus erythematosus, gout and Lyme arthritis, with a focus on the signalling mechanisms mediated by the Toll-IL-1 receptor (TIR) domain, the exogenous and endogenous ligands of TLRs, and the current and future therapeutic strategies to target TLR signalling in rheumatic diseases.
Collapse
|
45
|
Meriläinen L, Brander H, Herranen A, Schwarzbach A, Gilbert L. Pleomorphic forms of Borrelia burgdorferi induce distinct immune responses. Microbes Infect 2016; 18:484-95. [PMID: 27139815 DOI: 10.1016/j.micinf.2016.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 01/17/2016] [Accepted: 04/08/2016] [Indexed: 11/26/2022]
Abstract
Borrelia burgdorferi is the causative agent of tick-borne Lyme disease. As a response to environmental stress B. burgdorferi can change its morphology to a round body form. The role of B. burgdorferi pleomorphic forms in Lyme disease pathogenesis has long been debated and unclear. Here, we demonstrated that round bodies were processed differently in differentiated macrophages, consequently inducing distinct immune responses compared to spirochetes in vitro. Colocalization analysis indicated that the F-actin participates in internalization of both forms. However, round bodies end up less in macrophage lysosomes than spirochetes suggesting that there are differences in processing of these forms in phagocytic cells. Furthermore, round bodies stimulated distinct cytokine and chemokine production in these cells. We confirmed that spirochetes and round bodies present different protein profiles and antigenicity. In a Western blot analysis Lyme disease patients had more intense responses to round bodies when compared to spirochetes. These results suggest that round bodies have a role in Lyme disease pathogenesis.
Collapse
Affiliation(s)
- Leena Meriläinen
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland.
| | - Heini Brander
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland
| | - Anni Herranen
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland
| | | | - Leona Gilbert
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyvaskyla, P.O. Box 35, FI-40014 Jyvaskyla, Finland
| |
Collapse
|
46
|
Epigenetic regulation of neutrophil development and function. Semin Immunol 2016; 28:83-93. [PMID: 27084194 DOI: 10.1016/j.smim.2016.04.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/14/2022]
Abstract
In addition to performing well-defined effector functions, neutrophils are now recognized as versatile and sophisticated cells with critical immunoregulatory roles. These include the release of a variety of proinflammatory or immunosuppressive cytokines, as well as the expression of genes with regulatory functions. Neutrophils share broad transcriptional features with monocytes, in keeping with the close developmental relation between the two cell types. However, neutrophil-specific gene expression patterns conferring cell type-specific responses to bacterial, viral or fungal components have been identified. Accumulating evidence suggest that these differences reflect the peculiar epigenomic and regulatory landscapes of neutrophils and monocytes, in turn controlled by the specific lineage-determining transcription factors shaping their identity. In this review, we will describe current knowledge on how neutrophil identity and function are controlled at the molecular level, focusing on transcriptional and chromatin regulation of neutrophil development and activation in response to inflammatory stimuli.
Collapse
|
47
|
Shaw DK, Kotsyfakis M, Pedra JHF. For Whom the Bell Tolls (and Nods): Spit-acular Saliva. CURRENT TROPICAL MEDICINE REPORTS 2016; 3:40-50. [PMID: 27547699 DOI: 10.1007/s40475-016-0072-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Having emerged during the early part of the Cretaceous period, ticks are an ancient group of hematophagous ectoparasites with significant veterinary and public health importance worldwide. The success of their life strategy can be attributed, in part, to saliva. As we enter into a scientific era where the collection of massive data sets and structures for biological application is possible, we suggest that understanding the molecular mechanisms that govern the life cycle of ticks is within grasp. With this in mind, we discuss what is currently known regarding the manipulation of Toll-like (TLR) and Nod-like (NLR) receptor signaling pathways by tick salivary proteins, and how these molecules impact pathogen transmission.
Collapse
Affiliation(s)
- Dana K Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | - Michail Kotsyfakis
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Budweis, Czech Republic
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
48
|
Inhibition of TLR8 mediated signaling promotes BCG induced apoptosis in THP-1 cells. Microb Pathog 2015; 93:78-82. [PMID: 26657720 DOI: 10.1016/j.micpath.2015.11.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 01/16/2023]
Abstract
Apoptosis was considered as one of the important host defense mechanisms against mycobacteria infection. In macrophage, the main target cell of Mycobacterium tuberculosis, apoptosis after infection could help kill the bacillus inside and process the antigens for further presentation and proper immune response. Here, we identified a role of TLR8 during the apoptosis induced by Bacillus Calmette Guérin (BCG) infection in THP-1 cells. Knockdown TLR8 further increased the apoptosis induced by BCG infection, and this enhanced apoptosis was caspase-dependent. During this process, Erk1/2, JNK and NFκB pathways were negatively affected and contributed to the enhanced apoptosis.
Collapse
|
49
|
Meshkibaf S, Fritz J, Gottschalk M, Kim SO. Preferential production of G-CSF by a protein-like Lactobacillus rhamnosus GR-1 secretory factor through activating TLR2-dependent signaling events without activation of JNKs. BMC Microbiol 2015; 15:238. [PMID: 26502905 PMCID: PMC4623291 DOI: 10.1186/s12866-015-0578-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023] Open
Abstract
Background Different species and strains of probiotic bacteria confer distinct immunological responses on immune cells. Lactobacillus rhamnosus GR-1 (GR-1) is a probiotic bacterial strain found in both the intestinal and urogenital tracts, and has immunomodulatory effects on several cell types including macrophages. However, detailed immunological responses and the signaling mechanism involved in the response are largely unknown. Results We examined the production of GR-1-induced cytokines/chemokines and signaling events in macrophages. Among 84 cytokines and chemokines examined, GR-1 discretely induced granulocyte colony-stimulating factor (G-CSF) mRNA at highest levels (>60-fold) without inducing other cytokines such as IL-1α, IL-1β, IL-6 and TNF-α (<5-fold). The toll-like receptor (TLR) 2/6-agonist PAM2CSK4, TLR2/1-agonist PAM3CSK4 and TLR4-agonist lipopolysaccharide induced all of these inflammatory cytokines at high levels (>50-fold). The TLR2 ligand lipoteichoic acid activated all mitogen-activated kinases, Akt and NF-κB; whereas, GR-1 selectively activated extracellular regulated kinases and p38, NF-κB and Akt, but not c-Jun N-terminal kinases (JNKs) in a TLR2-dependent manner. Using specific inhibitors, we demonstrated that lack of JNKs activation by GR-1 caused inefficient production of pro-inflammatory cytokines but not G-CSF production. A secreted heat-labile protein-like molecule, 30–100 kDa in size, induced the preferential production of G-CSF. Conclusion This study elucidated unique signaling events triggered by GR-1, resulting in selective production of the immunomodulatory cytokine G-CSF in macrophages. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0578-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahab Meshkibaf
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, Western University, London, ON, N6G 2 V4, Canada. .,Center for Human Immunology, Western University, London, ON, N6G 2 V4, Canada.
| | - Jӧrg Fritz
- Department of Microbiology, McGill University, Montreal, QC, H3G 0B1, Canada.
| | - Marcelo Gottschalk
- Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC, J2S 2 M2, Canada.
| | - Sung Ouk Kim
- Department of Microbiology and Immunology and Infectious Diseases Research Group, Siebens-Drake Research Institute, Western University, London, ON, N6G 2 V4, Canada. .,Center for Human Immunology, Western University, London, ON, N6G 2 V4, Canada.
| |
Collapse
|
50
|
Bernard Q, Gallo RL, Jaulhac B, Nakatsuji T, Luft B, Yang X, Boulanger N. Ixodes tick saliva suppresses the keratinocyte cytokine response to TLR2/TLR3 ligands during early exposure to Lyme borreliosis. Exp Dermatol 2015; 25:26-31. [PMID: 26307945 DOI: 10.1111/exd.12853] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Ixodes hard tick induces skin injury by its sophisticated biting process. Its saliva plays a key role to enable an efficient blood meal that lasts for several days. We hypothesized that this feeding process may also be exploited by pathogens to facilitate their transmission, especially in the context of arthropod-borne diseases. To test this, we used Lyme borreliosis as a model. This bacterial infection is caused by Borrelia burgdorferi sensu lato transmitted by Ixodes. We co-incubated Borrelia with human keratinocytes in the presence of poly (I: C), a dsRNA TLR3 agonist generated by skin injury. This induced a strong cytokine response from human primary keratinocytes that was much greater than that induced by Borrelia alone. OspC, a TLR2/1 agonist and a major surface lipoprotein of Borrelia also amplified the process. Interestingly, tick saliva inhibited cytokine responses by keratinocytes to these TLR agonists. We propose that Borrelia uses the immunoprivileged site produced by tick saliva to facilitate its transmission.
Collapse
Affiliation(s)
- Quentin Bernard
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, CA, USA
| | - Benoît Jaulhac
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France.,Centre National de Reference Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, CA, USA
| | - Benjamin Luft
- Department of Medicine, State University of New York, Stony Brook, NY, USA
| | - Xiahoua Yang
- Department of Medicine, State University of New York, Stony Brook, NY, USA
| | - Nathalie Boulanger
- EA7290 Virulence bactérienne précoce: groupe borréliose de Lyme, Fédération de médecine Translationnelle et Faculté de Pharmacie de Strasbourg, Université de Strasbourg, Strasbourg, France.,Centre National de Reference Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| |
Collapse
|