1
|
Weerachatyanukul W, Pooljun C, Chotwiwatthanakun C, Jariyapong P. Binding of Infectious Hypodermal and Haematopoietic Necrosis Virus-Like Particles to Mannose Receptor Stimulates Antimicrobial Responses in Immune-Related Tissues of Peneaus vannamei. JOURNAL OF FISH DISEASES 2025; 48:e14051. [PMID: 39609717 DOI: 10.1111/jfd.14051] [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: 08/08/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024]
Abstract
Mannose receptor (MR) is a transmembrane protein and a type of pattern-recognition receptor (PRR) that plays a critical role in the immunity of mammals and fish. In this study, we examined the role of MR in binding with infectious hypodermal and haematopoietic necrosis virus-like particle (IHHN-VLP) and the downstream immune pathway that it triggers in the shrimp Peneaus vannamei. Upon IHHN-VLP challenge, transcripts of MR in P. vannamei (PvMR) increased significantly in all examined tissues, particularly those related to shrimp immunity, including hemocyte, hepatopancreas and gill tissues. Specifically, IHHN-VLP bound to the 34-kDa PvMR protein in shrimp-tissue extracts. Immunohistochemistry results of hemocytes showed that PvMR was initially localised on the plasma membrane but later internalised and dispersed throughout the cytoplasm after IHHN-VLP administration. Binding between IHHN-VLP and PvMR also induced significant upregulation of genes for the antimicrobial peptides (AMPs) penaeidin 3 and crustin, presumably to protect the shrimp against the viral infection. However, knocking down PvMR resulted in down-regulation of all immune-related genes examined. Overall, as an immune-related PRR, PvMR serves as a receptor for invading viruses, which then trigger the expression of AMPs. Strategic designs using PvMR could be developed to either block the interaction of native virus with the host cells or provoke its up-regulation to enhance shrimp immunity, which could open up opportunities to fight against IHHNV infection in shrimp.
Collapse
Affiliation(s)
| | - Chettupon Pooljun
- Akkhraratchakumari Veterinary College, Walailak University, Nakhonsrithammarat, Thailand
| | | | | |
Collapse
|
2
|
Khan S, Zuccato JA, Ignatchenko V, Singh O, Govindarajan M, Waas M, Mejia-Guerrero S, Gao A, Zadeh G, Kislinger T. Organelle resolved proteomics uncovers PLA2R1 as a novel cell surface marker required for chordoma growth. Acta Neuropathol Commun 2024; 12:39. [PMID: 38454495 PMCID: PMC10921702 DOI: 10.1186/s40478-024-01751-w] [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: 01/07/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024] Open
Abstract
Chordomas are clinically aggressive tumors with a high rate of disease progression despite maximal therapy. Given the limited therapeutic options available, there remains an urgent need for the development of novel therapies to improve clinical outcomes. Cell surface proteins are attractive therapeutic targets yet are challenging to profile with common methods. Four chordoma cell lines were analyzed by quantitative proteomics using a differential ultracentrifugation organellar fractionation approach. A subtractive proteomics strategy was applied to select proteins that are plasma membrane enriched. Systematic data integration prioritized PLA2R1 (secretory phospholipase A2 receptor-PLA2R1) as a chordoma-enriched surface protein. The expression profile of PLA2R1 was validated across chordoma cell lines, patient surgical tissue samples, and normal tissue lysates via immunoblotting. PLA2R1 expression was further validated by immunohistochemical analysis in a richly annotated cohort of 25-patient tissues. Immunohistochemistry analysis revealed that elevated expression of PLA2R1 is correlated with poor prognosis. Using siRNA- and CRISPR/Cas9-mediated knockdown of PLA2R1, we demonstrated significant inhibition of 2D, 3D and in vivo chordoma growth. PLA2R1 depletion resulted in cell cycle defects and metabolic rewiring via the MAPK signaling pathway, suggesting that PLA2R1 plays an essential role in chordoma biology. We have characterized the proteome of four chordoma cell lines and uncovered PLA2R1 as a novel cell-surface protein required for chordoma cell survival and association with patient outcome.
Collapse
Affiliation(s)
- Shahbaz Khan
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
| | - Jeffrey A Zuccato
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Vladimir Ignatchenko
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
| | - Olivia Singh
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
| | - Meinusha Govindarajan
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Matthew Waas
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
| | - Salvador Mejia-Guerrero
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
| | - Andrew Gao
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, Princess Margaret Cancer Research Tower, University Health Network, 101 College Street, Room 9-807, Toronto, ON, M5G 1L7, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| |
Collapse
|
3
|
Torrelles JB, Chatterjee D. Collected Thoughts on Mycobacterial Lipoarabinomannan, a Cell Envelope Lipoglycan. Pathogens 2023; 12:1281. [PMID: 38003746 PMCID: PMC10675199 DOI: 10.3390/pathogens12111281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
The presence of lipoarabinomannan (LAM) in the Mycobacterium tuberculosis (Mtb) cell envelope was first reported close to 100 years ago. Since then, numerous studies have been dedicated to the isolation, purification, structural definition, and elucidation of the biological properties of Mtb LAM. In this review, we present a brief historical perspective on the discovery of Mtb LAM and the herculean efforts devoted to structurally characterizing the molecule because of its unique structural and biological features. The significance of LAM remains high to this date, mainly due to its distinct immunological properties in conjunction with its role as a biomarker for diagnostic tests due to its identification in urine, and thus can serve as a point-of-care diagnostic test for tuberculosis (TB). In recent decades, LAM has been thoroughly studied and massive amounts of information on this intriguing molecule are now available. In this review, we give the readers a historical perspective and an update on the current knowledge of LAM with information on the inherent carbohydrate composition, which is unique due to the often puzzling sugar residues that are specifically found on LAM. We then guide the readers through the complex and myriad immunological outcomes, which are strictly dependent on LAM's chemical structure. Furthermore, we present issues that remain unresolved and represent the immediate future of LAM research. Addressing the chemistry, functions, and roles of LAM will lead to innovative ways to manipulate the processes that involve this controversial and fascinating biomolecule.
Collapse
Affiliation(s)
- Jordi B. Torrelles
- International Center for the Advancement of Research and Education (I • Care), Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Delphi Chatterjee
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
4
|
Gong Y, Wang J, Li F, Zhu B. Polysaccharides and glycolipids of Mycobacterium tuberculosis and their induced immune responses. Scand J Immunol 2023; 97:e13261. [PMID: 39008002 DOI: 10.1111/sji.13261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 07/16/2024]
Abstract
Tuberculosis (TB) is a chronic infectious disease mainly caused by Mycobacterium tuberculosis (M. tuberculosis). The structures of polysaccharides and glycolipids at M. tuberculosis cell wall vary among different strains, which affect the physiology and pathogenesis of mycobacteria by activating or inhibiting innate and acquired immunity. Among them, some components such as lipomannan (LM) and lipoarabinomannan (LAM) activate innate immunity by recognizing some kinds of pattern recognition receptors (PRRs) like Toll-like receptors, while other components like mannose-capped lipoarabinomannan (ManLAM) could prevent innate immune responses by inhibiting the secretion of pro-inflammatory cytokines and maturation of phagosomes. In addition, many glycolipids can activate natural killer T (NKT) cells and CD1-restricted T cells to produce interferon-γ (IFN-γ). Furthermore, humoral immunity against cell wall components, such as antibodies against LAM, plays a role in immunity against M. tuberculosis infection. Cell wall polysaccharides and glycolipids of M. tuberculosis have potential applications as antigens and adjuvants for novel TB subunit vaccines.
Collapse
Affiliation(s)
- Yang Gong
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Juan Wang
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Fei Li
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation & Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Correia-Neves M, Nigou J, Mousavian Z, Sundling C, Källenius G. Immunological hyporesponsiveness in tuberculosis: The role of mycobacterial glycolipids. Front Immunol 2022; 13:1035122. [PMID: 36544778 PMCID: PMC9761185 DOI: 10.3389/fimmu.2022.1035122] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 12/09/2022] Open
Abstract
Glycolipids constitute a major part of the cell envelope of Mycobacterium tuberculosis (Mtb). They are potent immunomodulatory molecules recognized by several immune receptors like pattern recognition receptors such as TLR2, DC-SIGN and Dectin-2 on antigen-presenting cells and by T cell receptors on T lymphocytes. The Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic relatives, phosphatidylinositol mannosides (PIMs) and lipomannan (LM), as well as other Mtb glycolipids, such as phenolic glycolipids and sulfoglycolipids have the ability to modulate the immune response, stimulating or inhibiting a pro-inflammatory response. We explore here the downmodulating effect of Mtb glycolipids. A great proportion of the studies used in vitro approaches although in vivo infection with Mtb might also lead to a dampening of myeloid cell and T cell responses to Mtb glycolipids. This dampened response has been explored ex vivo with immune cells from peripheral blood from Mtb-infected individuals and in mouse models of infection. In addition to the dampening of the immune response caused by Mtb glycolipids, we discuss the hyporesponse to Mtb glycolipids caused by prolonged Mtb infection and/or exposure to Mtb antigens. Hyporesponse to LAM has been observed in myeloid cells from individuals with active and latent tuberculosis (TB). For some myeloid subsets, this effect is stronger in latent versus active TB. Since the immune response in individuals with latent TB represents a more protective profile compared to the one in patients with active TB, this suggests that downmodulation of myeloid cell functions by Mtb glycolipids may be beneficial for the host and protect against active TB disease. The mechanisms of this downmodulation, including tolerance through epigenetic modifications, are only partly explored.
Collapse
Affiliation(s)
- Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal,Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B's), Portuguese (PT) Government Associate Laboratory, Braga, Portugal,Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier, Toulouse, France
| | - Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,School of Mathematics, Statistics, and Computer Science, College of Science, University of Tehran, Tehran, Iran,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gunilla Källenius
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,*Correspondence: Gunilla Källenius,
| |
Collapse
|
6
|
Fresquet M, Lockhart-Cairns MP, Rhoden SJ, Jowitt TA, Briggs DC, Baldock C, Brenchley PE, Lennon R. Structure of PLA2R reveals presentation of the dominant membranous nephropathy epitope and an immunogenic patch. Proc Natl Acad Sci U S A 2022; 119:e2202209119. [PMID: 35858348 PMCID: PMC9303975 DOI: 10.1073/pnas.2202209119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/31/2022] [Indexed: 01/04/2023] Open
Abstract
Membranous nephropathy is an autoimmune kidney disease caused by autoantibodies targeting antigens present on glomerular podocytes, instigating a cascade leading to glomerular injury. The most prevalent circulating autoantibodies in membranous nephropathy are against phospholipase A2 receptor (PLA2R), a cell surface receptor. The dominant epitope in PLA2R is located within the cysteine-rich domain, yet high-resolution structure-based mapping is lacking. In this study, we define the key nonredundant amino acids in the dominant epitope of PLA2R involved in autoantibody binding. We further describe two essential regions within the dominant epitope and spacer requirements for a synthetic peptide of the epitope for drug discovery. In addition, using cryo-electron microscopy, we have determined the high-resolution structure of PLA2R to 3.4 Å resolution, which shows that the dominant epitope and key residues within the cysteine-rich domain are accessible at the cell surface. In addition, the structure of PLA2R not only suggests a different orientation of domains but also implicates a unique immunogenic signature in PLA2R responsible for inducing autoantibody formation and recognition.
Collapse
Affiliation(s)
- Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Michael P. Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Samuel J. Rhoden
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Thomas A. Jowitt
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David C. Briggs
- Signalling and Structural Biology Lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Paul E. Brenchley
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
- Department of Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| |
Collapse
|
7
|
Lee S, Jang SH, Suzuki-Narita M, Gregoire S, Millecamps M, Stone LS. Voluntary running attenuates behavioural signs of low back pain: dimorphic regulation of intervertebral disc inflammation in male and female SPARC-null mice. Osteoarthritis Cartilage 2022; 30:110-123. [PMID: 34534663 DOI: 10.1016/j.joca.2021.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 05/12/2021] [Accepted: 06/18/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the effect of running exercise on behavioral measures of pain and intervertebral disc (IVD) inflammation in the SPARC-null mouse model. METHODS Male and female 8-month old SPARC-null and age-matched control mice received a home cage running wheel or a control, fixed wheel for 6 months. Behavioral assays were performed to assess axial discomfort (grip test) and radiating leg pain (von Frey, acetone tests) and voluntary running was confirmed. Expression of inflammatory mediators (TNF-α, IL-1β, IL-2, IL-10, CCL5, CXCL1, CXCL5, RANKL, M-CSF, and VEGF) in IVDs was determined. Additional inflammatory (IL-1β, IL-1Ra, CXCR1, CXCR2) and macrophage phenotypic markers (ITGAM, CD80, CD86, CD206, Arg1) in IVDs were investigated by qPCR. RESULTS Voluntary running attenuated behavioral measures of pain in male and female SPARC-null mice. Increases in mediators including IL-1β, CXCL1 and CXCL5 were observed in SPARC-null compared to control IVDs. After 6 months of running, increases in M-CSF and VEGF were observed in male SPARC-null IVDs. In females, pro-inflammatory mediators, including CXCL1 and CXCL5 were downregulated by running in SPARC-null mice. qPCR analysis further confirmed the anti-inflammatory effect of running in female IVDs with increased IL-1Ra mRNA. Running induced upregulation of the macrophage marker ITGAM mRNA in males. CONCLUSIONS Voluntary running reversed behavioral signs of pain in male and female mice and reduced inflammatory mediators in females, but not males. Thus, the therapeutic mechanism of action may be sex-specific.
Collapse
Affiliation(s)
- S Lee
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; The Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - S H Jang
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; The Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - M Suzuki-Narita
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - S Gregoire
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; The Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - M Millecamps
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; The Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - L S Stone
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; The Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada; Department of Anesthesiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Department of Pharmacology & Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Department of Neurology & Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Department of Anesthesiology, Faculty of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Single-molecule imaging of glycan-lectin interactions on cells with Glyco-PAINT. Nat Chem Biol 2021; 17:1281-1288. [PMID: 34764473 DOI: 10.1038/s41589-021-00896-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 09/10/2021] [Indexed: 11/08/2022]
Abstract
Most lectins bind carbohydrate ligands with relatively low affinity, making the identification of optimal ligands challenging. Here we introduce a point accumulation in nanoscale topography (PAINT) super-resolution microscopy method to capture weak glycan-lectin interactions at the single-molecule level in living cells (Glyco-PAINT). Glyco-PAINT exploits weak and reversible sugar binding to directly achieve single-molecule detection and quantification in cells and is used to establish the relative kon and koff rates of a synthesized library of carbohydrate-based probes, as well as the diffusion coefficient of the receptor-sugar complex. Uptake of ligands correlates with their binding affinity and residence time to establish structure-function relations for various synthetic glycans. We reveal how sugar multivalency and presentation geometry can be optimized for binding and internalization. Overall, Glyco-PAINT represents a powerful approach to study weak glycan-lectin interactions on the surface of living cells, one that can be potentially extended to a variety of lectin-sugar interactions.
Collapse
|
9
|
Holzheimer M, Buter J, Minnaard AJ. Chemical Synthesis of Cell Wall Constituents of Mycobacterium tuberculosis. Chem Rev 2021; 121:9554-9643. [PMID: 34190544 PMCID: PMC8361437 DOI: 10.1021/acs.chemrev.1c00043] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
The pathogen Mycobacterium tuberculosis (Mtb), causing
tuberculosis disease, features an extraordinary
thick cell envelope, rich in Mtb-specific lipids,
glycolipids, and glycans. These cell wall components are often directly
involved in host–pathogen interaction and recognition, intracellular
survival, and virulence. For decades, these mycobacterial natural
products have been of great interest for immunology and synthetic
chemistry alike, due to their complex molecular structure and the
biological functions arising from it. The synthesis of many of these
constituents has been achieved and aided the elucidation of their
function by utilizing the synthetic material to study Mtb immunology. This review summarizes the synthetic efforts of a quarter
century of total synthesis and highlights how the synthesis layed
the foundation for immunological studies as well as drove the field
of organic synthesis and catalysis to efficiently access these complex
natural products.
Collapse
Affiliation(s)
- Mira Holzheimer
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Jeffrey Buter
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
10
|
Ween MP, White JB, Tran HB, Mukaro V, Jones C, Macowan M, Hodge G, Trim PJ, Snel MF, Hodge SJ. The role of oxidised self-lipids and alveolar macrophage CD1b expression in COPD. Sci Rep 2021; 11:4106. [PMID: 33602992 PMCID: PMC7892841 DOI: 10.1038/s41598-021-82481-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
In chronic obstructive pulmonary disease (COPD) apoptotic bronchial epithelial cells are increased, and their phagocytosis by alveolar macrophages (AM) is decreased alongside bacterial phagocytosis. Epithelial cellular lipids, including those exposed on uncleared apoptotic bodies, can become oxidized, and may be recognized and presented as non-self by antigen presenting cells. CD1b is a lipid-presenting protein, previously only described in dendritic cells. We investigated whether CD1b is upregulated in COPD AM, and whether lipid oxidation products are found in the airways of cigarette smoke (CS) exposed mice. We also characterise CD1b for the first time in a range of macrophages and assess CD1b expression and phagocytic function in response to oxidised lipid. Bronchoalveolar lavage and exhaled breath condensate were collected from never-smoker, current-smoker, and COPD patients and AM CD1b expression and airway 8-isoprostane levels assessed. Malondialdehyde was measured in CS-exposed mouse airways by confocal/immunofluorescence. Oxidation of lipids produced from CS-exposed 16HBE14o- (HBE) bronchial epithelial cells was assessed by spectrophotometry and changes in lipid classes assessed by mass spectrometry. 16HBE cell toxicity was measured by flow cytometry as was phagocytosis, CD1b expression, HLA class I/II, and mannose receptor (MR) in monocyte derived macrophages (MDM). AM CD1b was significantly increased in COPD smokers (4.5 fold), COPD ex-smokers (4.3 fold), and smokers (3.9 fold), and AM CD1b significantly correlated with disease severity (FEV1) and smoking pack years. Airway 8-isoprostane also increased in smokers and COPD smokers and ex-smokers. Malondialdehyde was significantly increased in the bronchial epithelium of CS-exposed mice (MFI of 18.18 vs 23.50 for control). Oxidised lipid was produced from CS-exposed bronchial epithelial cells (9.8-fold of control) and showed a different overall lipid makeup to that of control total cellular lipid. This oxidised epithelial lipid significantly upregulated MDM CD1b, caused bronchial epithelial cell toxicity, and reduced MDM phagocytic capacity and MR in a dose dependent manner. Increased levels of oxidised lipids in the airways of COPD patients may be responsible for reduced phagocytosis and may become a self-antigen to be presented by CD1b on macrophages to perpetuate disease progression despite smoking cessation.
Collapse
Affiliation(s)
- Miranda P Ween
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia. .,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.
| | - Jake B White
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.,Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Hai B Tran
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Violet Mukaro
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Charles Jones
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Matthew Macowan
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Gregory Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Paul J Trim
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Marten F Snel
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Proteomics, Metabolomics and MS Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Sandra J Hodge
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
11
|
Prabhu S, Deng H, Cross TWL, Shahoei SH, Konopka CJ, Gonzalez Medina N, Applegate CC, Wallig MA, Dobrucki LW, Nelson ER, Smith AM, Swanson KS. Nanocarriers targeting adipose macrophages increase glucocorticoid anti-inflammatory potency to ameliorate metabolic dysfunction. Biomater Sci 2021; 9:506-518. [PMID: 33200765 DOI: 10.1039/d0bm01142h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity is associated with systemic inflammation due to macrophage accumulation in adipose tissue (AT). AT macrophages are, therefore, a target for therapeutics to modulate inflammation and prevent comorbidities. Because inflammatory processes have pleiotropic effects throughout the body and are intertwined with metabolic axes, systemic anti-inflammatory therapies are often harmful. We report that targeting AT macrophages using dextran nanocarriers radically alters the pharmacology of anti-inflammatory glucocorticoids, uncoupling the metabolic axis in obese mice. Following a single treatment, expression of inflammatory mediators and markers of inflammatory macrophages decreased with a nearly 20-fold higher potency compared with free drug. As a result, long-term treatment resulted in potent fat mobilization, AT reduction, weight loss, improved glucose tolerance, and altered AT gene expression profiles that led to elevated liver stress. Two weeks after treatment ceased, gene expression of inflammatory mediators in AT remained lower than obese controls, while gene expression related to metabolic function improved. These data demonstrate that nanocarriers show potential for amelioration of obesity-related AT inflammation and metabolic dysfunction, highlighting an important opportunity for nanomedicine to impact chronic metabolic disorders with complex and poorly understood etiology.
Collapse
Affiliation(s)
- Suma Prabhu
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Innate Immune Pattern Recognition Receptors of Mycobacterium tuberculosis: Nature and Consequences for Pathogenesis of Tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:179-215. [PMID: 34661896 DOI: 10.1007/978-3-030-67452-6_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Innate immunity against Mycobacterium tuberculosis is a critical early response to prevent the establishment of the infection. Despite recent advances in understanding the host-pathogen dialogue in the early stages of tuberculosis (TB), much has yet to be learnt. The nature and consequences of this dialogue ultimately determine the path of infection: namely, either early clearance of M. tuberculosis, or establishment of M. tuberculosis infection leading to active TB disease and/or latent TB infection. On the frontline in innate immunity are pattern recognition receptors (PRRs), with soluble factors (e.g. collectins and complement) and cell surface factors (e.g. Toll-like receptors and other C-type lectin receptors (Dectin 1/2, Nod-like receptors, DC-SIGN, Mincle, mannose receptor, and MCL) that play a central role in recognising M. tuberculosis and facilitating its clearance. However, in a 'double-edged sword' scenario, these factors can also be involved in enhancement of pathogenesis as well. Furthermore, innate immunity is also a critical bridge in establishing the subsequent adaptive immune response, which is also responsible for granuloma formation that cordons off M. tuberculosis infection, establishing latency and acting as a reservoir for bacterial persistence and dissemination of future disease. This chapter discusses the current understanding of pattern recognition of M. tuberculosis by innate immunity and the role this plays in the pathogenesis and protection against TB.
Collapse
|
13
|
Lu TM, Kanda M, Furuya H, Satoh N. Dicyemid Mesozoans: A Unique Parasitic Lifestyle and a Reduced Genome. Genome Biol Evol 2020; 11:2232-2243. [PMID: 31347665 PMCID: PMC6736024 DOI: 10.1093/gbe/evz157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2019] [Indexed: 12/25/2022] Open
Abstract
Dicyemids, previously called “mesozoans” (intermediates between unicellular protozoans and multicellular metazoans), are an enigmatic animal group. They have a highly simplified adult body, comprising only ∼30 cells, and they have a unique parasitic lifestyle. Recently, dicyemids were shown to be spiralians, with affinities to the Platyhelminthes. In order to understand molecular mechanisms involved in evolution of this odd animal, we sequenced the genome of Dicyema japonicum and a reference transcriptome assembly using mixed-stage samples. The D. japonicum genome features a high proportion of repetitive sequences that account for 49% of the genome. The dicyemid genome is reduced to ∼67.5 Mb with 5,012 protein-coding genes. Only four Hox genes exist in the genome, with no clustering. Gene distribution in KEGG pathways shows that D. japonicum has fewer genes in most pathways. Instead of eliminating entire critical metabolic pathways, parasitic lineages likely simplify pathways by eliminating pathway-specific genes, while genes with fundamental functions may be retained in multiple pathways. In principle, parasites can stand to lose genes that are unnecessary, in order to conserve energy. However, whether retained genes in incomplete pathways serve intermediate functions and how parasites overcome the physiological needs served by lost genes, remain to be investigated in future studies.
Collapse
Affiliation(s)
- Tsai-Ming Lu
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Japan.,Sars International Centre for Marine Molecular Biology, University of Bergen, Norway
| | - Miyuki Kanda
- DNA Sequencing Section, Okinawa Institute of Science and Technology Graduate University, Japan
| | - Hidetaka Furuya
- Department of Biology, Graduate School of Science, Osaka University, Japan
| | - Noriyuki Satoh
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Japan
| |
Collapse
|
14
|
Zhou KL, Li X, Zhang XL, Pan Q. Mycobacterial mannose-capped lipoarabinomannan: a modulator bridging innate and adaptive immunity. Emerg Microbes Infect 2019; 8:1168-1177. [PMID: 31379262 PMCID: PMC6713153 DOI: 10.1080/22221751.2019.1649097] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mannose-capped lipoarabinomannan (ManLAM) is a high molecular mass amphipathic lipoglycan identified in pathogenic Mycobacterium tuberculosis (M. tb) and M. bovis Bacillus Calmette-Guérin (BCG). ManLAM, serves as both an immunogen and a modulator of the host immune system, and its critical role in mycobacterial survival during infection has been well-characterized. ManLAM can be recognized by various types of receptors on both innate and adaptive immune cells, including macrophages, dendritic cells (DCs), neutrophils, natural killer T (NKT) cells, T cells and B cells. MamLAM has been shown to affect phagocytosis, cytokine production, antigen presentation, T cell activation and polarization, as well as antibody production. Exploring the mechanisms underlying the roles of ManLAM during mycobacterial infection will aid in improving tuberculosis (TB) prevention, diagnosis and treatment interventions. In this review, we highlight the interaction between ManLAM and receptors, intracellular signalling pathways triggered by ManLAM and its roles in both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Kai-Liang Zhou
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China.,b The eighth hospital of Wuhan , Wuhan , People's Republic of China
| | - Xin Li
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| | - Xiao-Lian Zhang
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| | - Qin Pan
- a State Key Laboratory of Virology and Medical Research Institue, Hubei Province Key Laboratory of Allergy and Immunology and Department of Immunology, Wuhan University School of Medicine , Wuhan , People's Republic of China
| |
Collapse
|
15
|
Jürgensen HJ, Silva LM, Krigslund O, van Putten S, Madsen DH, Behrendt N, Engelholm LH, Bugge TH. CCL2/MCP-1 signaling drives extracellular matrix turnover by diverse macrophage subsets. Matrix Biol Plus 2019; 1:100003. [PMID: 33543002 PMCID: PMC7852312 DOI: 10.1016/j.mbplus.2019.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Macrophage plasticity, cellular origin, and phenotypic heterogeneity are perpetual challenges for studies addressing the biology of this pivotal immune cell in development, homeostasis, and tissue remodeling/repair. Consequently, a myriad of macrophage subtypes has been described in these contexts. To facilitate the identification of functional macrophage subtypes in vivo, here we used a flow cytometry-based assay that allows for detailed phenotyping of macrophages engaged in extracellular matrix (ECM) degradation. Of the five macrophage subtypes identified in the remodeling dermis by using this assay, collagen degradation was primarily executed by Ly6C−CCR2+ and Ly6C−CCR2low macrophages via mannose receptor-dependent collagen endocytosis, while Ly6C+CCR2+ macrophages were the dominant fibrin-endocytosing cells. Unexpectedly, the CCL2/MCP1-CCR2 signaling axis was critical for both collagen and fibrin degradation, while collagen degradation was independent of IL-4Ra signaling. Furthermore, the cytokine GM-CSF selectively enhanced collagen degradation by Ly6C+CCR2+ macrophages. This study reveals distinct subsets of macrophages engaged in ECM turnover and identifies novel wound healing-associated functions for CCL2 and GM-CSF inflammatory cytokines. Phenotypically diverse subsets of dermal macrophages undertake the degradation of extracellular matrix C-C motif chemokine Ligand 2 (CCL2) signaling is critical for macrophage-mediated endocytosis of collagen and fibrin. Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and Interleukin (IL)-13 stimulate collagen endocytosis. The wound healing-associated IL4-IL4 Receptor a (IL4Ra) signaling is dispensable for collagen endocytosis by macrophages. The mannose receptor is the principal endocytic collagen receptor utilized by resident dermal macrophages.
Collapse
Key Words
- AF, Alexa Fluor
- CCL2/MCP-1, chemokine (C-C motif) ligand 2/monocyte chemoattractant protein 1
- CCR2, C-C chemokine receptor type 2
- CEMS, collagen-endocytosing macrophages
- Collagen degradation
- ECM, extracellular matrix
- Extracellular matrix endocytosis
- FEMS, fibrin-endocytosing macrophages
- FMO, fluorescence minus one
- Fibrin degradation
- GM-CSF, Granulocyte Macrophage-Colony Stimulating Factor
- GM-CSFR, GM-CSF Receptor
- IL, Interleukin
- IL4Ra, IL4 Receptor a
- Interleukin-13
- M-CSF, Macrophage-Colony Stimulating Factor
- MR, mannose receptor/CD206
- Mannose receptor/CD206
- Plg, plasminogen
- RFP, red fluorescent protein
- uPARAP, urokinase plasminogen activator receptor associated protein/Endo180
- uPARAP/Endo180
Collapse
Affiliation(s)
- Henrik J. Jürgensen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Lakmali M. Silva
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Oral Inflammation and Immunity Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Oliver Krigslund
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Sander van Putten
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Daniel H. Madsen
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
- Center for Cancer Immune Therapy (CCIT), Department of Haematology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
- Department of Oncology, Herlev Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Lars H. Engelholm
- Finsen Laboratory, Rigshospitalet/BRIC, University of Copenhagen, Ole Maaloesvej 5, DK-2200 Copenhagen N, Denmark
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
- Corresponding author at: Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 211, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Hu Z, Shi X, Yu B, Li N, Huang Y, He Y. Structural Insights into the pH-Dependent Conformational Change and Collagen Recognition of the Human Mannose Receptor. Structure 2017; 26:60-71.e3. [PMID: 29225077 DOI: 10.1016/j.str.2017.11.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/14/2017] [Accepted: 11/08/2017] [Indexed: 11/28/2022]
Abstract
Mannose receptor (MR, CD206) is an endocytic receptor on microphages and dendritic cells. It recognizes multiple ligands and plays important roles in regulating immune responses and maintaining glycoprotein homeostasis. However, the structure and functional mechanism of MR remain unclear. Here we determine the crystal structures of the N-terminal fragments of MR and reveal the potential binding mode of collagen on the fibronectin II domain. The SAXS and other biophysical data suggest that MR adopts an extended conformation at physiological pH and undergoes conformational changes as pH decreases, resulting in a compact conformation in an acidic environment. Moreover, biochemical data show that MR binds to collagen in a Ca2+-enhanced manner at physiological pH, whereas Ca2+ has no effect on the binding at acidic pH. These results provide a model for the dynamic mechanism of MR regarding its ligand binding and release during the recycling between cell surface and endosomes.
Collapse
Affiliation(s)
- Zhenzheng Hu
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China
| | - Xiangyi Shi
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China
| | - Bowen Yu
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China
| | - Na Li
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China
| | - Ying Huang
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China
| | - Yongning He
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China.
| |
Collapse
|
17
|
Moliva JI, Turner J, Torrelles JB. Immune Responses to Bacillus Calmette-Guérin Vaccination: Why Do They Fail to Protect against Mycobacterium tuberculosis? Front Immunol 2017; 8:407. [PMID: 28424703 PMCID: PMC5380737 DOI: 10.3389/fimmu.2017.00407] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis (TB), is the current leading cause of death due to a single infectious organism. Although curable, the broad emergence of multi-, extensive-, extreme-, and total-drug resistant strains of M.tb has hindered eradication efforts of this pathogen. Furthermore, computational models predict a quarter of the world’s population is infected with M.tb in a latent state, effectively serving as the largest reservoir for any human pathogen with the ability to cause significant morbidity and mortality. The World Health Organization has prioritized new strategies for improved vaccination programs; however, the lack of understanding of mycobacterial immunity has made it difficult to develop new successful vaccines. Currently, Mycobacterium bovis bacillus Calmette–Guérin (BCG) is the only vaccine approved for use to prevent TB. BCG is highly efficacious at preventing meningeal and miliary TB, but is at best 60% effective against the development of pulmonary TB in adults and wanes as we age. In this review, we provide a detailed summary on the innate immune response of macrophages, dendritic cells, and neutrophils in response to BCG vaccination. Additionally, we discuss adaptive immune responses generated by BCG vaccination, emphasizing their specific contributions to mycobacterial immunity. The success of future vaccines against TB will directly depend on our understanding of mycobacterial immunity.
Collapse
Affiliation(s)
- Juan I Moliva
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
18
|
Orr SL, Tobias P. LPS and LAM activation of the U373 astrocytoma cell line: Differential requirement for CD14. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060030201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD14 is a membrane protein (mCD14) found on monocytes and neutrophils that is required for the innate immune response to lipopolysaccharide (LPS) and lipoarabinomannan (LAM). CD14 can also be found in serum as soluble CD14 (sCD14) that when bound to bacterial products, enables many non-CD14 bearing cells to be activated. Lipopolysaccharide binding protein (LBP) is a plasma protein that disaggregates and catalytically transfers LPS to CD14. To examine the role of CD14 and LBP in LAM-dependent activation, we used the U373 astrocyte cell line to stably express membrane-bound CD14 (U373-CD14). In serum-free conditions, U373-CD14 cells could respond to LAM stimulation as measured by expression of intracellular adhesion molecule-1 (ICAM-1). Vector control cells (U373-RSV) could not respond to LAM or LPS; but, upon the addition of serum as a source of soluble CD14, control U373-RSV cells could respond to LPS, but not LAM. Therefore, LAM can activate U373 cells only through membrane CD14 and not soluble CD14. We also demonstrate that this membrane CD14-dependent LAM response is greatly facilitated by the addition of LBP.
Collapse
Affiliation(s)
- Sally L. Orr
- The Scripps Research Institute, La Jolla, California, USA,
| | - Peter Tobias
- The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
19
|
Huang S, Moody DB. Donor-unrestricted T cells in the human CD1 system. Immunogenetics 2016; 68:577-96. [PMID: 27502318 PMCID: PMC5915868 DOI: 10.1007/s00251-016-0942-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/14/2016] [Indexed: 02/06/2023]
Abstract
The CD1 and MHC systems are specialized for lipid and peptide display, respectively. Here, we review evidence showing how cellular CD1a, CD1b, CD1c, and CD1d proteins capture and display many cellular lipids to T cell receptors (TCRs). Increasing evidence shows that CD1-reactive T cells operate outside two classical immunogenetic concepts derived from the MHC paradigm. First, because CD1 proteins are non-polymorphic in human populations, T cell responses are not restricted to the donor's genetic background. Second, the simplified population genetics of CD1 antigen-presenting molecules can lead to simplified patterns of TCR usage. As contrasted with donor-restricted patterns of MHC-TCR interaction, the donor-unrestricted nature of CD1-TCR interactions raises the prospect that lipid agonists and antagonists of T cells could be developed.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - D Branch Moody
- Divison of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
20
|
C-type lectin receptors in tuberculosis: what we know. Med Microbiol Immunol 2016; 205:513-535. [DOI: 10.1007/s00430-016-0470-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022]
|
21
|
Franken L, Schiwon M, Kurts C. Macrophages: sentinels and regulators of the immune system. Cell Microbiol 2016; 18:475-87. [PMID: 26880038 DOI: 10.1111/cmi.12580] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 12/15/2022]
Abstract
The important role of macrophages in host defense against a variety of pathogens has long been recognized and has been documented and reviewed in numerous publications. Recently, it has become clear that tissue macrophages are not entirely derived from monocytes, as has been assumed for a long time, but rather show an ontogenetic dichotomy in most tissues: while part of the tissue macrophages are derived from monocytes, a major subset is prenatally seeded from the yolk sac. The latter subset shows a remarkable longevity and is maintained by self-renewal in the adult animal. This paradigm shift poses interesting questions: are these two macrophage subsets functionally equivalent cells that are recruited into the tissue at different development stages, or are both macrophage subsets discrete cell types with distinct functions, which have to exist side by side? Is the functional specialization that can be observed in most macrophages due to their lineage or due to their anatomical niche? This review will give an overview about what we know of macrophage ontogeny and will discuss the influence of the macrophage lineage and location on their functional specialization.
Collapse
Affiliation(s)
- Lars Franken
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| | - Marzena Schiwon
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-University, Sigmund-Freud-Str. 25, Bonn, 53105, Germany
| |
Collapse
|
22
|
McIntosh JD, Brimble MA, Brooks AES, Dunbar PR, Kowalczyk R, Tomabechi Y, Fairbanks AJ. Convergent chemo-enzymatic synthesis of mannosylated glycopeptides; targeting of putative vaccine candidates to antigen presenting cells. Chem Sci 2015; 6:4636-4642. [PMID: 28717478 PMCID: PMC5500846 DOI: 10.1039/c5sc00952a] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/11/2015] [Indexed: 01/11/2023] Open
Abstract
The combination of solid phase peptide synthesis and endo-β-N-acetylglucosaminidase (ENGase) catalysed glycosylation is a powerful convergent synthetic method allowing access to glycopeptides bearing full-length N-glycan structures. Mannose-terminated N-glycan oligosaccharides, produced by either total or semi-synthesis, were converted into oxazoline donor substrates. A peptide from the human cytomegalovirus (CMV) tegument protein pp65 that incorporates a well-characterised T cell epitope, containing N-acetylglucosamine at specific Asn residues, was accessed by solid phase peptide synthesis, and used as an acceptor substrate. High-yielding enzymatic glycosylation afforded glycopeptides bearing defined homogeneous high-mannose N-glycan structures. These high-mannose containing glycopeptides were tested for enhanced targeting to human antigen presenting cells (APCs), putatively mediated via the mannose receptor, and for processing by the APCs for presentation to human CD8+ T cells specific for a 9-mer epitope within the peptide. Binding assays showed increased binding of glycopeptides to APCs compared to the non-glycosylated control. Glycopeptides bearing high-mannose N-glycan structures at a single site outside the T cell epitope were processed and presented by the APCs to allow activation of a T cell clone. However, the addition of a second glycan within the T cell epitope resulted in ablation of T cell activation. We conclude that chemo-enzymatic synthesis of mannosylated glycopeptides enhances uptake by human APCs while preserving the immunogenicity of peptide epitopes within the glycopeptides, provided those epitopes are not themselves glycosylated.
Collapse
Affiliation(s)
- Julie D McIntosh
- School of Biological Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences , The University of Auckland , 23 Symonds St , Auckland , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - Anna E S Brooks
- School of Biological Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - P Rod Dunbar
- School of Biological Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - Renata Kowalczyk
- School of Chemical Sciences , The University of Auckland , 23 Symonds St , Auckland , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - Yusuke Tomabechi
- Department of Chemistry , University of Canterbury , Private Bag 4800 , Christchurch , 8140 , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| | - Antony J Fairbanks
- Department of Chemistry , University of Canterbury , Private Bag 4800 , Christchurch , 8140 , New Zealand .
- Maurice Wilkins Centre for Molecular Biodiscovery , University of Auckland , Private Bag 92019 , Auckland 1010 , New Zealand
| |
Collapse
|
23
|
Salio M, Cerundolo V. Regulation of Lipid Specific and Vitamin Specific Non-MHC Restricted T Cells by Antigen Presenting Cells and Their Therapeutic Potentials. Front Immunol 2015; 6:388. [PMID: 26284072 PMCID: PMC4517378 DOI: 10.3389/fimmu.2015.00388] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/13/2015] [Indexed: 12/17/2022] Open
Abstract
Since initial reports, more than 25 years ago, that T cells recognize lipids in the context on non-polymorphic CD1 molecules, our understanding of antigen presentation to non-peptide-specific T cell populations has deepened. It is now clear that αβ T cells bearing semi-invariant T cell receptor, as well as subsets of γδ T cells, recognize a variety of self and non-self lipids and contribute to shaping immune responses via cross talk with dendritic cells and B cells. Furthermore, it has been demonstrated that small molecules derived from the microbial riboflavin biosynthetic pathway (vitamin B2) bind monomorphic MR1 molecules and activate mucosal-associated invariant T cells, another population of semi-invariant T cells. Novel insights in the biological relevance of non-peptide-specific T cells have emerged with the development of tetrameric CD1 and MR1 molecules, which has allowed accurate enumeration and functional analysis of CD1- and MR1-restricted T cells in humans and discovery of novel populations of semi-invariant T cells. The phenotype and function of non-peptide-specific T cells will be discussed in the context of the known distribution of CD1 and MR1 molecules by different subsets of antigen-presenting cells at steady state and following infection. Concurrent modulation of CD1 transcription and lipid biosynthetic pathways upon TLR stimulation, coupled with efficient lipid antigen processing, result in the increased cell surface expression of antigenic CD1-lipid complexes. Similarly, MR1 expression is almost undetectable in resting APC and it is upregulated following bacterial infection, likely due to stabilization of MR1 molecules by microbial antigens. The tight regulation of CD1 and MR1 expression at steady state and during infection may represent an important mechanism to limit autoreactivity, while promoting T cell responses to foreign antigens.
Collapse
Affiliation(s)
- Mariolina Salio
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Kallert S, Zenk SF, Walther P, Grieshober M, Weil T, Stenger S. Liposomal delivery of lipoarabinomannan triggers Mycobacterium tuberculosis specific T-cells. Tuberculosis (Edinb) 2015; 95:452-62. [PMID: 26043674 DOI: 10.1016/j.tube.2015.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 12/20/2022]
Abstract
Lipoarabinomannan (LAM) is a major cell wall component of Mycobacterium tuberculosis (Mtb). LAM specific human T-lymphocytes release interferon-γ (IFNγ) and have antimicrobial activity against intracellular Mtb suggesting that they contribute to protection. Therefore the induction of LAM-specific memory T-cells is an attractive approach for the design of a new vaccine against tuberculosis. A prerequisite for the activation of LAM-specific T-cells is the efficient uptake and transport of the glycolipid antigen to the CD1 antigen presenting machinery. Based on the hydrophobicity of LAM we hypothesized that packaging of LAM into liposomes will support the activation of T-lymphocytes. We prepared liposomes containing phosphatidylcholine, cholesterol, stearylated octaarginine and LAM via thin layer hydration method (LIPLAM). Flow cytometry analysis using fluorescently labelled LIPLAM showed an efficient uptake by antigen presenting cells. LAM delivered via liposomes was biologically active as demonstrated by the down-regulation of peroxisome proliferator activated receptor gamma (PPARγ) protein expression. Importantly, LIPLAM induced higher IFNγ production by primary human T-lymphocytes than purified LAM (2-16 times) or empty liposomes. These results suggest that the delivery of mycobacterial glycolipids via liposomes is a promising approach to promote the induction of M. tuberculosis specific T-cell responses.
Collapse
Affiliation(s)
- Stephanie Kallert
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Sebastian F Zenk
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Paul Walther
- Central Unit Electron Microscopy, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Mark Grieshober
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| | - Tanja Weil
- Institute for Organic Chemistry III/Macromolecular Chemistry, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Albert Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
25
|
Jones CH, Gollakota A, Chen M, Chung TC, Ravikrishnan A, Zhang G, Pfeifer BA. Influence of molecular weight upon mannosylated bio-synthetic hybrids for targeted antigen presenting cell gene delivery. Biomaterials 2015; 58:103-11. [PMID: 25941787 DOI: 10.1016/j.biomaterials.2015.04.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 11/24/2022]
Abstract
Given the rise of antibiotic resistant microbes, genetic vaccination is a promising prophylactic strategy that enables rapid design and manufacture. Facilitating this process is the choice of vector, which is often situationally-specific and limited in engineering capacity. Furthermore, these shortcomings are usually tied to an incomplete understanding of the structure-function relationships driving vector-mediated gene delivery. Building upon our initial report of a hybrid bacterial-biomaterial gene delivery vector, a comprehensive structure-function assessment was completed using a class of mannosylated poly(beta-amino esters). Through a top-down screening methodology, an ideal polymer was selected on the basis of gene delivery efficacy and then used for the synthesis of a stratified molecular weight polymer library. By eliminating contributions of polymer chemical background, we were able to complete an in-depth assessment of gene delivery as a function of (1) polymer molecular weight, (2) relative mannose content, (3) polymer-membrane biophysical properties, (4) APC uptake specificity, and (5) serum inhibition. In summary, the flexibility and potential of the hybrid design featured in this work highlights the ability to systematically probe vector-associated properties for the development of translational gene delivery candidates.
Collapse
Affiliation(s)
- Charles H Jones
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Akhila Gollakota
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Mingfu Chen
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Tai-Chun Chung
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Anitha Ravikrishnan
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Guojian Zhang
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA
| | - Blaine A Pfeifer
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260-4200, USA.
| |
Collapse
|
26
|
Abstract
For decades, proteins were thought to be the sole or at least the dominant source of antigens for T cells. Studies in the 1990s demonstrated that CD1 proteins and mycobacterial lipids form specific targets of human αβ T cells. The molecular basis by which T-cell receptors (TCRs) recognize CD1-lipid complexes is now well understood. Many types of mycobacterial lipids function as antigens in the CD1 system, and new studies done with CD1 tetramers identify T-cell populations in the blood of tuberculosis patients. In human populations, a fundamental difference between the CD1 and major histocompatibility complex systems is that all humans express nearly identical CD1 proteins. Correspondingly, human CD1 responsive T cells show evidence of conserved TCRs. In addition to natural killer T cells and mucosal-associated invariant T (MAIT cells), conserved TCRs define other subsets of human T cells, including germline-encoded mycolyl-reactive (GEM) T cells. The simple immunogenetics of the CD1 system and new investigative tools to measure T-cell responses in humans now creates a situation in which known lipid antigens can be developed as immunodiagnostic and immunotherapeutic reagents for tuberculosis disease.
Collapse
Affiliation(s)
- Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
27
|
Staines K, Hunt LG, Young JR, Butter C. Evolution of an expanded mannose receptor gene family. PLoS One 2014; 9:e110330. [PMID: 25390371 PMCID: PMC4229073 DOI: 10.1371/journal.pone.0110330] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 09/21/2014] [Indexed: 01/22/2023] Open
Abstract
Sequences of peptides from a protein specifically immunoprecipitated by an antibody, KUL01, that recognises chicken macrophages, identified a homologue of the mammalian mannose receptor, MRC1, which we called MRC1L-B. Inspection of the genomic environment of the chicken gene revealed an array of five paralogous genes, MRC1L-A to MRC1L-E, located between conserved flanking genes found either side of the single MRC1 gene in mammals. Transcripts of all five genes were detected in RNA from a macrophage cell line and other RNAs, whose sequences allowed the precise definition of spliced exons, confirming or correcting existing bioinformatic annotation. The confirmed gene structures were used to locate orthologues of all five genes in the genomes of two other avian species and of the painted turtle, all with intact coding sequences. The lizard genome had only three genes, one orthologue of MRC1L-A and two orthologues of the MRC1L-B antigen gene resulting from a recent duplication. The Xenopus genome, like that of most mammals, had only a single MRC1-like gene at the corresponding locus. MRC1L-A and MRC1L-B genes had similar cytoplasmic regions that may be indicative of similar subcellular migration and functions. Cytoplasmic regions of the other three genes were very divergent, possibly indicating the evolution of a new functional repertoire for this family of molecules, which might include novel interactions with pathogens.
Collapse
Affiliation(s)
| | | | | | - Colin Butter
- The Pirbright Institute, Compton, United Kingdom
| |
Collapse
|
28
|
Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol 2014; 26:471-85. [PMID: 25453226 DOI: 10.1016/j.smim.2014.09.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022]
Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Claire E Dodd
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
29
|
Azad AK, Rajaram MVS, Schlesinger LS. Exploitation of the Macrophage Mannose Receptor (CD206) in Infectious Disease Diagnostics and Therapeutics. ACTA ACUST UNITED AC 2014; 1. [PMID: 24672807 DOI: 10.13188/2325-4653.1000003] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The macrophage mannose receptor (MR, CD206) is a C-type lectin expressed predominantly by most tissue macrophages, dendritic cells and specific lymphatic or endothelial cells. It functions in endocytosis and phagocytosis, and plays an important role in immune homeostasis by scavenging unwanted mannoglycoproteins. More attention is being paid to its particularly high expression in tissue pathology sites during disease such the tumor microenvironment. The MR recognizes a variety of microorganisms by their mannan-coated cell wall, which is exploited by adapted intracellular pathogens such as Mycobacterium tuberculosis, for their own survival. Despite the continued development of drug delivery technologies, the targeting of agents to immune cells, especially macrophages, for effective diagnosis and treatment of chronic infectious diseases has not been addressed adequately. In this regard, strategies that optimize MR-mediated uptake by macrophages in target tissues during infection are becoming an attractive approach. We review important progress in this area.
Collapse
Affiliation(s)
- Abul K Azad
- Center for Microbial Interface Biology and the Department of Microbial Infection and Immunity, The Ohio State University, USA
| | - Murugesan V S Rajaram
- Center for Microbial Interface Biology and the Department of Microbial Infection and Immunity, The Ohio State University, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology and the Department of Microbial Infection and Immunity, The Ohio State University, USA
| |
Collapse
|
30
|
Targeting antigens to dendritic cell receptors for vaccine development. JOURNAL OF DRUG DELIVERY 2013; 2013:869718. [PMID: 24228179 PMCID: PMC3817681 DOI: 10.1155/2013/869718] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/11/2013] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) are highly specialized antigen presenting cells of the immune system which play a key role in regulating immune responses. Depending on the method of antigen delivery, DCs stimulate immune responses or induce tolerance. As a consequence of the dual function of DCs, DCs are studied in the context of immunotherapy for both cancer and autoimmune diseases. In vaccine development, a major aim is to induce strong, specific T-cell responses. This is achieved by targeting antigen to cell surface molecules on DCs that efficiently channel the antigen into endocytic compartments for loading onto MHC molecules and stimulation of T-cell responses. The most attractive cell surface receptors, expressed on DCs used as targets for antigen delivery for cancer and other diseases, are discussed.
Collapse
|
31
|
Madsen DH, Leonard D, Masedunskas A, Moyer A, Jürgensen HJ, Peters DE, Amornphimoltham P, Selvaraj A, Yamada SS, Brenner DA, Burgdorf S, Engelholm LH, Behrendt N, Holmbeck K, Weigert R, Bugge TH. M2-like macrophages are responsible for collagen degradation through a mannose receptor-mediated pathway. J Cell Biol 2013; 202:951-66. [PMID: 24019537 PMCID: PMC3776354 DOI: 10.1083/jcb.201301081] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 07/31/2013] [Indexed: 02/07/2023] Open
Abstract
Tissue remodeling processes critically depend on the timely removal and remodeling of preexisting collagen scaffolds. Nevertheless, many aspects related to the turnover of this abundant extracellular matrix component in vivo are still incompletely understood. We therefore took advantage of recent advances in optical imaging to develop an assay to visualize collagen turnover in situ and identify cell types and molecules involved in this process. Collagen introduced into the dermis of mice underwent cellular endocytosis in a partially matrix metalloproteinase-dependent manner and was subsequently routed to lysosomes for complete degradation. Collagen uptake was predominantly executed by a quantitatively minor population of M2-like macrophages, whereas more abundant Col1a1-expressing fibroblasts and Cx3cr1-expressing macrophages internalized collagen at lower levels. Genetic ablation of the collagen receptors mannose receptor (Mrc1) and urokinase plasminogen activator receptor-associated protein (Endo180 and Mrc2) impaired this intracellular collagen degradation pathway. This study demonstrates the importance of receptor-mediated cellular uptake to collagen turnover in vivo and identifies a key role of M2-like macrophages in this process.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- CX3C Chemokine Receptor 1
- Cell Proliferation
- Cells, Cultured
- Collagen/metabolism
- Collagen Type I/physiology
- Collagen Type I, alpha 1 Chain
- Endocytosis/physiology
- Female
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Immunoenzyme Techniques
- Lysosomes/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Cell Surface/physiology
- Receptors, Chemokine/physiology
- Receptors, Immunologic
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
Collapse
Affiliation(s)
- Daniel H. Madsen
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Daniel Leonard
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Andrius Masedunskas
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Amanda Moyer
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Henrik Jessen Jürgensen
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Diane E. Peters
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- Program of Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Panomwat Amornphimoltham
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Arul Selvaraj
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Susan S. Yamada
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - David A. Brenner
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sven Burgdorf
- Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Lars H. Engelholm
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Niels Behrendt
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Kenn Holmbeck
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Roberto Weigert
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section and Intracellular Membrane Trafficking Unit, Oral and Pharyngeal Cancer Branch, and Matrix Metalloproteinase Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
32
|
Macrophages in tuberculosis: friend or foe. Semin Immunopathol 2013; 35:563-83. [PMID: 23864058 DOI: 10.1007/s00281-013-0388-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/20/2013] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis (Mtb), is acquired by the respiratory route. It is exquisitely human adapted and a prototypic intracellular pathogen of macrophages, with alveolar macrophages (AMs) being the primary conduit of infection and disease. The outcome of primary infection is most often a latently infected healthy human host, in whom the bacteria are held in check by the host immune response. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the host immune response fails to control the growth of bacilli, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols coughed out into the environment and inhaled by new hosts. The molecular details of the Mtb-macrophage interaction continue to be elucidated. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. Macrophages demonstrate tremendous phenotypic heterogeneity and functional plasticity which, depending on the site and stage of infection, facilitate the diverse outcomes. Moreover, host responses vary depending on the specific characteristics of the infecting Mtb strain. In this chapter, we describe a contemporary view of the behavior of AMs and their interaction with various Mtb strains in generating unique immunologic lung-specific responses.
Collapse
|
33
|
Yang CY, Chen JB, Tsai TF, Tsai YC, Tsai CY, Liang PH, Hsu TL, Wu CY, Netea MG, Wong CH, Hsieh SL. CLEC4F is an inducible C-type lectin in F4/80-positive cells and is involved in alpha-galactosylceramide presentation in liver. PLoS One 2013; 8:e65070. [PMID: 23762286 PMCID: PMC3675125 DOI: 10.1371/journal.pone.0065070] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 04/21/2013] [Indexed: 01/19/2023] Open
Abstract
CLEC4F, a member of C-type lectin, was first purified from rat liver extract with high binding affinity to fucose, galactose (Gal), N-acetylgalactosamine (GalNAc), and un-sialylated glucosphingolipids with GalNAc or Gal terminus. However, the biological functions of CLEC4F have not been elucidated. To address this question, we examined the expression and distribution of murine CLEC4F, determined its binding specificity by glycan array, and investigated its function using CLEC4F knockout (Clec4f-/-) mice. We found that CLEC4F is a heavily glycosylated membrane protein co-expressed with F4/80 on Kupffer cells. In contrast to F4/80, CLEC4F is detectable in fetal livers at embryonic day 11.5 (E11.5) but not in yolk sac, suggesting the expression of CLEC4F is induced as cells migrate from yolk cells to the liver. Even though CLEC4F is not detectable in tissues outside liver, both residential Kupffer cells and infiltrating mononuclear cells surrounding liver abscesses are CLEC4F-positive upon Listeria monocytogenes (L. monocytogenes) infection. While CLEC4F has strong binding to Gal and GalNAc, terminal fucosylation inhibits CLEC4F recognition to several glycans such as Fucosyl GM1, Globo H, Bb3∼4 and other fucosyl-glycans. Moreover, CLEC4F interacts with alpha-galactosylceramide (α-GalCer) in a calcium-dependent manner and participates in the presentation of α-GalCer to natural killer T (NKT) cells. This suggests that CLEC4F is a C-type lectin with diverse binding specificity expressed on residential Kupffer cells and infiltrating monocytes in the liver, and may play an important role to modulate glycolipids presentation on Kupffer cells.
Collapse
Affiliation(s)
- Chih-Ya Yang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jiun-Bo Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chen Tsai
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Ching-Yen Tsai
- Transgenic Core Facility, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Clinical Medicine & Infection and Immunity Center, National Yang-Ming University, Taipei, Taiwan
- Immunology Center, Taipei Veterans General Hospital, Taipei, Taiwan
- The Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
34
|
Tjomsland V, Ellegård R, Burgener A, Mogk K, Che KF, Westmacott G, Hinkula J, Lifson JD, Larsson M. Complement opsonization of HIV-1 results in a different intracellular processing pattern and enhanced MHC class I presentation by dendritic cells. Eur J Immunol 2013; 43:1470-83. [PMID: 23526630 PMCID: PMC3738931 DOI: 10.1002/eji.201242935] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 02/20/2013] [Accepted: 03/19/2013] [Indexed: 11/11/2022]
Abstract
Induction of optimal HIV-1-specific T-cell responses, which can contribute to controlling viral infection in vivo, depends on antigen processing and presentation processes occurring in DCs. Opsonization can influence the routing of antigen processing and pathways used for presentation. We studied antigen proteolysis and the role of endocytic receptors in MHC class I (MHCI) and II (MHCII) presentation of antigens derived from HIV-1 in human monocyte-derived immature DCs (IDCs) and mature DCs, comparing free and complement opsonized HIV-1 particles. Opsonization of virions promoted MHCI presentation by DCs, indicating that complement opsonization routes more virions toward the MHCI presentation pathway. Blockade of macrophage mannose receptor (MMR) and β7-integrin enhanced MHCI and MHCII presentation by IDCs and mature DCs, whereas the block of complement receptor 3 decreased MHCI and MHCII presentation. In addition, we found that IDC and MDC proteolytic activities were modulated by HIV-1 exposure; complement-opsonized HIV-1 induced an increased proteasome activity in IDCs. Taken together, these findings indicate that endocytic receptors such as MMR, complement receptor 3, and β7-integrin can promote or disfavor antigen presentation probably by routing HIV-1 into different endosomal compartments with distinct efficiencies for degradation of viral antigens and MHCI and MHCII presentation, and that HIV-1 affects the antigen-processing machinery.
Collapse
Affiliation(s)
- Veronica Tjomsland
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköping, Sweden
| | - Rada Ellegård
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköping, Sweden
| | - Adam Burgener
- Department of Medical Microbiology, University of ManitobaWinnipeg, Canada
| | - Kenzie Mogk
- Department of Medical Microbiology, University of ManitobaWinnipeg, Canada
| | - Karlhans F Che
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköping, Sweden
| | | | - Jorma Hinkula
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköping, Sweden
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory for Cancer ResearchFrederick, MD, USA
| | - Marie Larsson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköping, Sweden
| |
Collapse
|
35
|
Van Rhijn I, Ly D, Moody DB. CD1a, CD1b, and CD1c in immunity against mycobacteria. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:181-97. [PMID: 23468110 DOI: 10.1007/978-1-4614-6111-1_10] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The CD1 system is composed of five types of human CD1 proteins, CD1a, CD1b, CD1c, CD1d, and CD1e, and their mammalian orthologs. Each type of CD1 protein has a distinct antigen binding groove and shows differing patterns of expression within cells and in different tissues. Here we review the molecular mechanisms by which CD1a, CD1b, and CD1c capture distinct classes of self- and mycobacterial antigens. We discuss how CD1-restricted T cells participate in the immune response, emphasizing new evidence for mycobacterial recognition in vivo in human and non-human models.
Collapse
Affiliation(s)
- Ildiko Van Rhijn
- Division of Rheumatology, Harvard Medical School, Boston, MA, USA.
| | | | | |
Collapse
|
36
|
Duluc D, Gannevat J, Joo H, Ni L, Upchurch K, Boreham M, Carley M, Stecher J, Zurawski G, Oh S. Dendritic cells and vaccine design for sexually-transmitted diseases. Microb Pathog 2012. [PMID: 23201532 DOI: 10.1016/j.micpath.2012.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are major antigen presenting cells (APCs) that can initiate and control host immune responses toward either immunity or tolerance. These features of DCs, as immune orchestrators, are well characterized by their tissue localizations as well as by their subset-dependent functional specialties and plasticity. Thus, the level of protective immunity to invading microbial pathogens can be dependent on the subsets of DCs taking up microbial antigens and their functional plasticity in response to microbial products, host cellular components and the cytokine milieu in the microenvironment. Vaccines are the most efficient and cost-effective preventive medicine against infectious diseases. However, major challenges still remain for the diseases caused by sexually-transmitted pathogens, including HIV, HPV, HSV and Chlamydia. We surmise that the establishment of protective immunity in the female genital mucosa, the major entry and transfer site of these pathogens, will bring significant benefit for the protection against sexually-transmitted diseases. Recent progresses made in DC biology suggest that vaccines designed to target proper DC subsets may permit us to establish protective immunity in the female genital mucosa against sexually-transmitted pathogens.
Collapse
Affiliation(s)
- Dorothee Duluc
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Afonso-Barroso A, Clark SO, Williams A, Rosa GT, Nóbrega C, Silva-Gomes S, Vale-Costa S, Ummels R, Stoker N, Movahedzadeh F, van der Ley P, Sloots A, Cot M, Appelmelk BJ, Puzo G, Nigou J, Geurtsen J, Appelberg R. Lipoarabinomannan mannose caps do not affect mycobacterial virulence or the induction of protective immunity in experimental animal models of infection and have minimal impact on in vitro inflammatory responses. Cell Microbiol 2012; 15:660-74. [PMID: 23121245 DOI: 10.1111/cmi.12065] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 10/24/2012] [Accepted: 10/28/2012] [Indexed: 11/27/2022]
Abstract
Mannose-capped lipoarabinomannan (ManLAM) is considered an important virulence factor of Mycobacterium tuberculosis. However, while mannose caps have been reported to be responsible for various immunosuppressive activities of ManLAM observed in vitro, there is conflicting evidence about their contribution to mycobacterial virulence in vivo. Therefore, we used Mycobacterium bovis BCG and M. tuberculosis mutants that lack the mannose cap of LAM to assess the role of ManLAM in the interaction of mycobacteria with the host cells, to evaluate vaccine-induced protection and to determine its importance in M. tuberculosis virulence. Deletion of the mannose cap did not affect BCG survival and replication in macrophages, although the capless mutant induced a somewhat higher production of TNF. In dendritic cells, the capless mutant was able to induce the upregulation of co-stimulatory molecules and the only difference we detected was the secretion of slightly higher amounts of IL-10 as compared to the wild type strain. In mice, capless BCG survived equally well and induced an immune response similar to the parental strain. Furthermore, the efficacy of vaccination against a M. tuberculosis challenge in low-dose aerosol infection models in mice and guinea pigs was not affected by the absence of the mannose caps in the BCG. Finally, the lack of the mannose cap in M. tuberculosis did not affect its virulence in mice nor its interaction with macrophages in vitro. Thus, these results do not support a major role for the mannose caps of LAM in determining mycobacterial virulence and immunogenicity in vivo in experimental animal models of infection, possibly because of redundancy of function.
Collapse
|
38
|
Abstract
Tuberculosis (TB) is a leading cause worldwide of human mortality attributable to a single infectious agent. Recent studies targeting candidate genes and "case-control" association have revealed numerous polymorphisms implicated in host susceptibility to TB. Here, we review current progress in the understanding of causative polymorphisms in host innate immune genes associated with TB pathogenesis. We discuss genes encoding several types of proteins: macrophage receptors, such as the mannose receptor (MR, CD206), dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN, CD209), Dectin-1, Toll-like receptors (TLRs), complement receptor 3 (CR3, CD11b/CD18), nucleotide oligomerization domain 1 (NOD1) and NOD2, CD14, P2X7, and the vitamin D nuclear receptor (VDR); soluble C-type lectins, such as surfactant protein-A (SP-A), SP-D, and mannose-binding lectin (MBL); phagocyte cytokines, such as tumor necrosis factor (TNF), interleukin-1β (IL-1β), IL-6, IL-10, IL-12, and IL-18; chemokines, such as IL-8, monocyte chemoattractant protein 1 (MCP-1), RANTES, and CXCL10; and other important innate immune molecules, such as inducible nitric oxide synthase (iNOS) and solute carrier protein 11A1 (SLC11A1). Polymorphisms in these genes have been variably associated with susceptibility to TB among different populations. This apparent variability is probably accounted for by evolutionary selection pressure as a result of long-term host-pathogen interactions in certain regions or populations and, in part, by lack of proper study design and limited knowledge of molecular and functional effects of the implicated genetic variants. Finally, we discuss genomic technologies that hold promise for resolving questions regarding the evolutionary paths of the human genome, functional effects of polymorphisms, and corollary impacts of adaptation on human health, ultimately leading to novel approaches to controlling TB.
Collapse
Affiliation(s)
- Abul K. Azad
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology
| | - Wolfgang Sadee
- Department of Pharmacology, Program in Pharmacogenomics, The Ohio State University, Columbus, Ohio, USA
| | - Larry S. Schlesinger
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology
| |
Collapse
|
39
|
Vigerust DJ, Vick S, Shepherd VL. Characterization of functional mannose receptor in a continuous hybridoma cell line. BMC Immunol 2012; 13:51. [PMID: 22967244 PMCID: PMC3495026 DOI: 10.1186/1471-2172-13-51] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 08/30/2012] [Indexed: 12/29/2022] Open
Abstract
Background The mannose receptor is the best described member of the type I transmembrane C-type lectins; however much remains unanswered about the biology of the receptor. One difficulty has been the inability to consistently express high levels of a functional full length mannose receptor cDNA in mammalian cells. Another difficulty has been the lack of a human macrophage cell line expressing a fully functional receptor. Commonly used human macrophage cell lines such as U937, THP-1, Mono-Mac and HL60 do not express the mannose receptor. We have developed a macrophage hybridoma cell line (43MR cells) created by fusion of U937 cells with primary human monocyte-derived macrophages, resulting in a non-adherent cell line expressing several properties of primary macrophages. The purpose of this study was to identify and select mannose receptor-expressing cells using fluorescence-activated cell sorting and to characterize the expression and function of the receptor. Results In the current study we show that the mannose receptor found on this novel cell has endocytic characteristics consistent with and similar to the mannose receptor found on the surface of monocyte-derived human macrophages and rat bone marrow-derived macrophages. In addition, we demonstrate that these cells engage and internalize pathogen particles such as S. aureus and C. albicans. We further establish the transfectability of these cells via the introduction of a plasmid expressing influenza A hemagglutinin. Conclusions The 43MR cell line represents the first naturally expressed MR-positive cell line derived from a human macrophage background. This cell line provides an important cell model for other researchers for the study of human MR biology and host-pathogen interactions.
Collapse
Affiliation(s)
- David J Vigerust
- Department of Veterans Affairs Medical Center, VA Medical Center/Research Service, 1310 24th Ave,, South, Nashville TN 37212, USA
| | | | | |
Collapse
|
40
|
Torrelles JB, Sieling PA, Zhang N, Keen MA, McNeil MR, Belisle JT, Modlin RL, Brennan PJ, Chatterjee D. Isolation of a distinct Mycobacterium tuberculosis mannose-capped lipoarabinomannan isoform responsible for recognition by CD1b-restricted T cells. Glycobiology 2012; 22:1118-27. [PMID: 22534567 PMCID: PMC3382347 DOI: 10.1093/glycob/cws078] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/17/2012] [Accepted: 04/19/2012] [Indexed: 12/23/2022] Open
Abstract
Mannose-capped lipoarabinomannan (ManLAM) is a complex lipoglycan abundantly present in the Mycobacterium tuberculosis cell envelope. Many biological properties have been ascribed to ManLAM, from directly interacting with the host and participating in the intracellular survival of M. tuberculosis, to triggering innate and adaptive immune responses, including the activation of CD1b-restricted T cells. Due to its structural complexity, ManLAM is considered a heterogeneous population of molecules which may explain its different biological properties. The presence of various modifications such as fatty acids, succinates, lactates, phosphoinositides and methylthioxylose in ManLAM have proven to correlate directly with its biological activity and may potentially be involved in the interactions between CD1b and the T cell population. To further delineate the specific ManLAM epitopes involved in CD1b-restricted T cell recognition, and their potential roles in mediating immune responses in M. tuberculosis infection, we established a method to resolve ManLAM into eight different isoforms based on their different isoelectric values. Our results show that a ManLAM isoform with an isoelectric value of 5.8 was the most potent in stimulating the production of interferon-γ in different CD1b-restricted T-cell lines. Compositional analyses of these isoforms of ManLAM revealed a direct relationship between the overall charge of the ManLAM molecule and its capacity to be presented to T cells via the CD1 compartment.
Collapse
Affiliation(s)
- Jordi B Torrelles
- Mycobacterial Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Abstract
ADAMTS13 is a plasma metalloproteinase that regulates platelet adhesion and aggregation by cleaving ultra-large VWF multimers on the surfaces of endothelial cells. Autoantibodies directed against ADAMTS13 prohibit the processing of VWF multimers, initiating a rare and life-threatening disorder called acquired thrombotic thrombocytopenic purpura. The formation of autoantibodies depends on the activation of CD4(+) T cells. This process requires immune recognition, endocytosis, and subsequent processing of ADAMTS13 into peptides that are presented on MHC class II molecules to CD4(+) T cells by dendritic cells (DCs). In the present study, we investigated endocytosis of recombinant ADAMTS13 by immature monocyte-derived DCs using flow cytometry and confocal microscopy. After incubation of fluorescently labeled ADAMTS13 with DCs, significant uptake of ADAMTS13 was observed. Endocytosis of ADAMTS13 was completely blocked by the addition of EGTA and mannan. ADAMTS13 endocytosis was decreased in the presence of a blocking mAb directed toward the macrophage mannose receptor (MR). Furthermore, siRNA silencing of MR reduced the uptake of ADAMTS13 by DCs. In addition, in vitro binding studies confirmed the interaction of ADAMTS13 with the carbohydrate recognition domains of MR. The results of the present study indicate that sugar moieties on ADAMTS13 interact with MR, thereby promoting its endocytosis by APCs.
Collapse
|
43
|
Huang S, Cheng TY, Young DC, Layre E, Madigan CA, Shires J, Cerundolo V, Altman JD, Moody DB. Discovery of deoxyceramides and diacylglycerols as CD1b scaffold lipids among diverse groove-blocking lipids of the human CD1 system. Proc Natl Acad Sci U S A 2011; 108:19335-40. [PMID: 22087000 PMCID: PMC3228429 DOI: 10.1073/pnas.1112969108] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Unlike the dominant role of one class II invariant chain peptide (CLIP) in blocking MHC class II, comparative lipidomics analysis shows that human cluster of differentiation (CD) proteins CD1a, CD1b, CD1c, and CD1d bind lipids corresponding to hundreds of diverse accurate mass retention time values. Although most ions were observed in association with several CD1 proteins, ligands binding selectively to one CD1 isoform allowed the study of how differing antigen-binding grooves influence lipid capture. Although the CD1b groove is distinguished by its unusually large volume (2,200 Å(3)) and the T' tunnel, the average mass of compounds eluted from CD1b was similar to that of lipids from CD1 proteins with smaller grooves. Elution of small ligands from the large CD1b groove might be explained if two small lipids bind simultaneously in the groove. Crystal structures indicate that all CD1 proteins can capture one antigen with its hydrophilic head group exposed for T-cell recognition, but CD1b structures show scaffold lipids seated below the antigen. We found that ligands selectively associated with CD1b lacked the hydrophilic head group that is generally needed for antigen recognition but interferes with scaffold function. Furthermore, we identified the scaffolds as deoxyceramides and diacylglycerols and directly demonstrate a function in augmenting presentation of a small glycolipid antigen to T cells. Thus, unlike MHC class II, CD1 proteins capture highly diverse ligands in the secretory pathway. CD1b has a mechanism for presenting either two small or one large lipid, allowing presentation of antigens with an unusually broad range of chain lengths.
Collapse
Affiliation(s)
- Shouxiong Huang
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Tan-Yun Cheng
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - David C. Young
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Emilie Layre
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Cressida A. Madigan
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - John Shires
- Emory Vaccine Center, Emory School of Medicine, Atlanta, GA 30322; and
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - John D. Altman
- Emory Vaccine Center, Emory School of Medicine, Atlanta, GA 30322; and
| | - D. Branch Moody
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
44
|
Londrigan SL, Tate MD, Brooks AG, Reading PC. Cell-surface receptors on macrophages and dendritic cells for attachment and entry of influenza virus. J Leukoc Biol 2011; 92:97-106. [PMID: 22124137 PMCID: PMC7166464 DOI: 10.1189/jlb.1011492] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Review of interactions between influenza A virus and C‐type lectin receptors on macrophages and dendritic cells that may result in virus entry and infection. Airway MΦ and DCs are important components of innate host defense and can play a critical role in limiting the severity of influenza virus infection. Although it has been well established that cell‐surface SA acts as a primary attachment receptor for IAV, the particular receptor(s) or coreceptor(s) that mediate IAV entry into any cell, including MΦ and DC, have not been clearly defined. Identifying which receptors are involved in attachment and entry of IAV into immune cells may have important implications in regard to understanding IAV tropism and pathogenesis. Recent evidence suggests that specialized receptors on MΦ and DCs, namely CLRs, can act as capture and/or entry receptors for many viral pathogens, including IAV. Herein, we review the early stages of infection of MΦ and DC by IAV. Specifically, we examine the potential role of CLRs expressed on MΦ and DC to act as attachment and/or entry receptors for IAV.
Collapse
Affiliation(s)
- Sarah L Londrigan
- The Department of Microbiology and Immunology, The University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
45
|
Garrido VV, Dulgerian LR, Stempin CC, Cerbán FM. The increase in mannose receptor recycling favors arginase induction and Trypanosoma cruzi survival in macrophages. Int J Biol Sci 2011; 7:1257-72. [PMID: 22110379 PMCID: PMC3221363 DOI: 10.7150/ijbs.7.1257] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 01/10/2023] Open
Abstract
The macrophage mannose receptor (MR) is a pattern recognition receptor of the innate immune system that binds to microbial structures bearing mannose, fucose and N-acetylglucosamine on their surface. Trypanosoma cruzi antigen cruzipain (Cz) is found in the different developmental forms of the parasite. This glycoprotein has a highly mannosylated C-terminal domain that participates in the host-antigen contact. Our group previously demonstrated that Cz-macrophage (Mo) interaction could modulate the immune response against T. cruzi through the induction of a preferential metabolic pathway. In this work, we have studied in Mo the role of MR in arginase induction and in T. cruzi survival using different MR ligands. We have showed that pre-incubation of T. cruzi infected cells with mannose-Bovine Serum Albumin (Man-BSA, MR specific ligand) biased nitric oxide (NO)/urea balance towards urea production and increased intracellular amastigotes growth. The study of intracellular signals showed that pre-incubation with Man-BSA in T. cruzi J774 infected cells induced down-regulation of JNK and p44/p42 phosphorylation and increased of p38 MAPK phosphorylation. These results are coincident with previous data showing that Cz also modifies the MAPK phosphorylation profile induced by the parasite. In addition, we have showed by confocal microscopy that Cz and Man-BSA enhance MR recycling. Furthermore, we studied MR behavior during T. cruzi infection in vivo. MR was up-regulated in F4/80+ cells from T. cruzi infected mice at 13 and 15 days post infection. Besides, we investigated the effect of MR blocking antibody in T. cruzi infected peritoneal Mo. Arginase activity and parasite growth were decreased in infected cells pre-incubated with anti-MR antibody as compared with infected cells treated with control antibody. Therefore, we postulate that during T. cruzi infection, Cz may contact with MR, increasing MR recycling which leads to arginase activity up-regulation and intracellular parasite growth.
Collapse
Affiliation(s)
- Vanina V Garrido
- CIBICI-CONICET, Dpto Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | | | | | | |
Collapse
|
46
|
Torrelles JB, Sieling PA, Arcos J, Knaup R, Bartling C, Rajaram MVS, Stenger S, Modlin RL, Schlesinger LS. Structural differences in lipomannans from pathogenic and nonpathogenic mycobacteria that impact CD1b-restricted T cell responses. J Biol Chem 2011; 286:35438-35446. [PMID: 21859718 DOI: 10.1074/jbc.m111.232587] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mannosylated molecules on the Mycobacterium tuberculosis surface are important determinants in the immunopathogenesis of tuberculosis. To date, much attention has been paid to mannose-capped lipoarabinomannan, which mediates phagocytosis and intracellular trafficking of M. tuberculosis by engaging the macrophage mannose receptor and subsequently binds to intracellular CD1b molecules for presentation to T cells. Another important mannosylated lipoglycan on the M. tuberculosis surface is lipomannan (LM). Comparative structural detail of the LMs from virulent and avirulent strains is limited as is knowledge regarding their differential capacity to be recognized by the adaptive immune response. Here, we purified LM from the avirulent M. smegmatis and the virulent M. tuberculosis H(37)R(v), performed a comparative structural biochemical analysis, and addressed their ability to stimulate CD1b-restricted T cell clones. We found that M. tuberculosis H(37)R(v) produces a large neutral LM (TB-LM); in contrast, M. smegmatis produces a smaller linear acidic LM (SmegLM) with a high succinate content. Correspondingly, TB-LM was not as efficiently presented to CD1b-restricted T cells as SmegLM. Thus, here we correlate the structure-function relationships for LMs with CD1b-restricted T cell responses and provide evidence that the structural features of TB-LM contribute to its diminished T cell responsiveness.
Collapse
Affiliation(s)
- Jordi B Torrelles
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Peter A Sieling
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Jesús Arcos
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Rose Knaup
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Craig Bartling
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Murugesan V S Rajaram
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital of Ulm, D-89081 Ulm, Germany
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Larry S Schlesinger
- Center for Microbial Interface Biology and Departments of Microbial Infection and Immunity and Internal Medicine, Ohio State University, Columbus, Ohio 43210.
| |
Collapse
|
47
|
Mishra AK, Driessen NN, Appelmelk BJ, Besra GS. Lipoarabinomannan and related glycoconjugates: structure, biogenesis and role in Mycobacterium tuberculosis physiology and host-pathogen interaction. FEMS Microbiol Rev 2011; 35:1126-57. [PMID: 21521247 PMCID: PMC3229680 DOI: 10.1111/j.1574-6976.2011.00276.x] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Approximately one third of the world's population is infected with Mycobacterium tuberculosis, the causative agent of tuberculosis. This bacterium has an unusual lipid-rich cell wall containing a vast repertoire of antigens, providing a hydrophobic impermeable barrier against chemical drugs, thus representing an attractive target for vaccine and drug development. Apart from the mycolyl–arabinogalactan–peptidoglycan complex, mycobacteria possess several immunomodulatory constituents, notably lipomannan and lipoarabinomannan. The availability of whole-genome sequences of M. tuberculosis and related bacilli over the past decade has led to the identification and functional characterization of various enzymes and the potential drug targets involved in the biosynthesis of these glycoconjugates. Both lipomannan and lipoarabinomannan possess highly variable chemical structures, which interact with different receptors of the immune system during host–pathogen interactions, such as Toll-like receptors-2 and C-type lectins. Recently, the availability of mutants defective in the synthesis of these glycoconjugates in mycobacteria and the closely related bacterium, Corynebacterium glutamicum, has paved the way for host–pathogen interaction studies, as well as, providing attenuated strains of mycobacteria for the development of new vaccine candidates. This review provides a comprehensive account of the structure, biosynthesis and immunomodulatory properties of these important glycoconjugates.
Collapse
Affiliation(s)
- Arun K Mishra
- School of Biosciences, University of Birmingham, Edgbaston, UK
| | | | | | | |
Collapse
|
48
|
Torrelles JB, Schlesinger LS. Diversity in Mycobacterium tuberculosis mannosylated cell wall determinants impacts adaptation to the host. Tuberculosis (Edinb) 2010; 90:84-93. [PMID: 20199890 PMCID: PMC2855779 DOI: 10.1016/j.tube.2010.02.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 02/05/2010] [Indexed: 10/19/2022]
Abstract
Mycobacterium tuberculosis (the causal agent of TB) has co-evolved with humans for centuries. It infects via the airborne route and is a prototypic highly adapted intracellular pathogen of macrophages. Extensive sequencing of the M. tuberculosis genome along with recent molecular phylogenetic studies is enabling us to gain insight into the biologic diversity that exists among bacterial strains that impact the pathogenesis of latent infection and disease. The majority of the M. tuberculosis cell envelope is comprised of carbohydrates and lipids, and there is increasing evidence that these microbial determinants that are readily exposed to the host immune system play critical roles in disease pathogenesis. Studies from our laboratory and others have raised the possibility that M. tuberculosis is adapting to the human host by cloaking its cell envelope molecules with terminal mannosylated (i.e. Man-alpha-(1-->2)-Man) oligosaccharides that resemble the glycoforms of mammalian mannoproteins. These mannosylated biomolecules engage the mannose receptor (MR) on macrophages during phagocytosis and dictate the intracellular fate of M. tuberculosis by regulating formation of the unique vesicular compartment in which the bacterium survives. The MR is highly expressed on alveolar macrophages (predominant C-type lectin on human cells) and functions as a scavenger receptor to maintain the healthiness of the lung by clearing foreign particles and at the same time regulating dangerous inflammatory responses. Thus M. tuberculosis exploits MR functions to gain entry into the macrophage and survive. Key biochemical pathways and mycobacterial determinants involved in the development and maintenance of the M. tuberculosis phagosome are being identified. The phylogenetic diversity observed in M. tuberculosis strains that impact its cell wall structure together with the genetic diversity observed in human populations, including those elements that affect macrophage function, may help to explain the extraordinary evolutionary adaptation of this pathogen to the human host. Major developments in these areas are the focus of this review.
Collapse
Affiliation(s)
- Jordi B Torrelles
- Center for Microbial Interface Biology, Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | | |
Collapse
|
49
|
Cernadas M, Cavallari M, Watts G, Mori L, De Libero G, Brenner MB. Early recycling compartment trafficking of CD1a is essential for its intersection and presentation of lipid antigens. THE JOURNAL OF IMMUNOLOGY 2009; 184:1235-41. [PMID: 20026739 DOI: 10.4049/jimmunol.0804140] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A major step in understanding differences in the nature of Ag presentation was the realization that MHC class I samples peptides transported to the endoplasmic reticulum from the cytosol, whereas MHC class II samples peptides from lysosomes. In contrast to MHC class I and II molecules that present protein Ags, CD1 molecules present lipid Ags for recognition by specific T cells. Each of the five members of the CD1 family (CD1a-e) localizes to a distinct subcompartment of endosomes. Accordingly, it has been widely assumed that the distinct trafficking of CD1 isoforms must also have evolved to enable them to sample lipid Ags that traffic via different routes. Among the CD1 isoforms, CD1a is unusual because it does not have a tyrosine-based cytoplasmic sorting motif and uniquely localizes to the early endocytic recycling compartment. This led us to predict that CD1a might have evolved to focus on lipids that localize to early endocytic/recycling compartments. Strikingly, we found that the glycolipid Ag sulfatide also localized almost exclusively to early endocytic and recycling compartments. Consistent with colocalization of CD1a and sulfatide, wild-type CD1a molecules efficiently presented sulfatide to CD1a-restricted, sulfatide-specific T cells. In contrast, CD1a:CD1b tail chimeras, that retain the same Ag-binding capacity as CD1a but traffic based on the cytoplasmic tail of CD1b to lysosomes, failed to present sulfatide efficiently. Thus, the intracellular trafficking route of CD1a is essential for efficient presentation of lipid Ags that traffic through the early endocytic and recycling pathways.
Collapse
Affiliation(s)
- Manuela Cernadas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
50
|
Soluble CD36 ectodomain binds negatively charged diacylglycerol ligands and acts as a co-receptor for TLR2. PLoS One 2009; 4:e7411. [PMID: 19847289 PMCID: PMC2760212 DOI: 10.1371/journal.pone.0007411] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 09/18/2009] [Indexed: 11/19/2022] Open
Abstract
Background Cluster of differentiation 36 (CD36) is a transmembrane glycoprotein involved in many biological processes, such as platelet biology, angiogenesis and in the aetiopathology of atherosclerosis and cardiovascular diseases. Toll-like receptors (TLRs) are one of the most important receptors of the innate immune system. Their main function is the recognition of conserved structure of microorganisms. This recognition triggers signaling pathways that activate transcription of cytokines and co-stimulatory molecules which participate in the generation of an immune response against microbes. In particular, TLR2 has been shown to recognize a broad range of ligands. Recently, we showed that CD36 serves as a co-receptor for TLR2 and enhances recognition of specific diacylglycerides derived from bacteria. Methodology/ Principal Findings Here, we investigate the mechanism by which CD36 contributes to ligand recognition and activation of TLR2 signaling pathway. We show that the ectodomain of murine CD36 (mCD36ED) directly interacts with negatively charged diacylglycerol ligands, which explains the specificity and selectivity of CD36 as a TLR2 co-receptor. We also show that mCD36ED amplifies the pro-inflammatory response to lipoteichoic acid in macrophages of wild-type mice and restores the pro-inflammatory response of macrophages from mice deficient in CD36 (oblivious), but not from mice deficient in cluster of differentiation 14 (CD14) (heedless). Conclusion/ Significance These data indicate that the CD36 ectodomain is the only relevant domain for activation of TLR2 signaling pathway and that CD36 and CD14 have a non-redundant role for loading ligands onto TLR2 in the plasma-membrane. The pro-inflammatory role of soluble CD36 can be relevant in the activation of the immune response against pathogens, as well as in the progression of chronic diseases. Therefore, an increased level of soluble forms of CD36, which has been reported to be increased in type II diabetic patients, could accelerate atherosclerosis by increasing the pro-inflammatory response to diacylglycerol ligands.
Collapse
|