1
|
Rungelrath V, Ahmed M, Hicks L, Miller SM, Ryter KT, Montgomery K, Ettenger G, Riffey A, Abdelwahab WM, Khader SA, Evans JT. Vaccination with Mincle agonist UM-1098 and mycobacterial antigens induces protective Th1 and Th17 responses. NPJ Vaccines 2024; 9:100. [PMID: 38844494 PMCID: PMC11156909 DOI: 10.1038/s41541-024-00897-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is one of the top infectious killers in the world. The only licensed vaccine against TB, Bacille Calmette-Guérin (BCG), provides variable protection against pulmonary TB, especially in adults. Hence, novel TB vaccine approaches are urgently needed. Both Th1 and Th17 responses are necessary for protection against TB, yet effective adjuvants and vaccine delivery systems for inducing robust Th1 and Th17 immunity are lacking. Herein we describe a synthetic Mincle agonist, UM-1098, and a silica nanoparticle delivery system that drives Th1/Th17 responses to Mtb antigens. Stimulation of human peripheral blood mononuclear cells (hPBMCs) with UM-1098 induced high levels of Th17 polarizing cytokines IL-6, IL-1β, IL-23 as well as IL-12p70, IL-4 and TNF-α in vitro. PBMCs from both C57BL/6 and BALB/c mice responded with a similar cytokine pattern in vitro and in vivo. Importantly, intramuscular (I.M.) vaccination with UM-1098-adjuvanted TB antigen M72 resulted in significantly higher antigen-specific IFN-γ and IL-17A levels in C57BL/6 wt mice than Mincle KO mice. Vaccination of C57BL/6 wt mice with immunodominant Mtb antigens ESAT6/Ag85B or M72 resulted in predominantly Th1 and Th17 responses and induced antigen-specific serum antibodies. Notably, in a virulent Mtb challenge model, vaccination with UM-1098 adjuvanted ESAT6/Ag85B or M72 significantly reduced lung bacterial burden when compared with unvaccinated mice and protection occurred in the absence of pulmonary inflammation. These data demonstrate that the synthetic Mincle agonist UM-1098 induces strong Th1 and Th17 immunity after vaccination with Mtb antigens and provides protection against Mtb infection in mice.
Collapse
Affiliation(s)
- Viktoria Rungelrath
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Linda Hicks
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kendal T Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kyle Montgomery
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - George Ettenger
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Shabaana Abdul Khader
- Department of Microbiology, University of Chicago, 920 E. 58th St., Chicago, IL, 60637, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA.
- Department of Biomedical & Pharmaceutical Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
2
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
3
|
Zhou C, Cai S, Xie Y, Zeng Z, Zhang J, Su G, Wu Q, Ye X, Cao Q, Yang P, Hu J. Genetic association of PRKCD and CARD9 polymorphisms with Vogt-Koyanagi-Harada disease in the Chinese Han population. Hum Genomics 2023; 17:9. [PMID: 36782298 PMCID: PMC9926551 DOI: 10.1186/s40246-023-00459-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Protein kinase C delta (PRKCD) and caspase recruitment domain family member 9 (CARD9) are genes involved in B and T cell activation, and cytokine production, which are vital mechanisms underlying autoimmune disease development. This study aimed to explore the association of the PRKCD and CARD9 genes with Vogt-Koyanagi-Harada disease (VKH) disease. The case-control study was performed to in 912 patients with VKH and 878 normal controls. MassARRAY system, SHEsis online platform, real-time PCR, and enzyme-linked immunosorbent assay were used to detect genotyping, haplotyping, mRNA expression, and cytokine levels, respectively. RESULTS We found that rs74437127 C allele of PRKCD, rs3812555 CC genotype, and C allele of CARD9 were associated with increased susceptibility of VKH (Pc = 0.020, OR = 1.624; Pc = 2.04 × 10-5, OR = 1.810; Pc = 2.76 × 10-5, OR = 1.698, respectively). However, the rs74437127 T allele, and rs3812555 TC genotype and T allele were linked with decreased susceptibility to VKH (Pc = 0.020, OR = 0.616; Pc = 7.85 × 10-5, OR = 0.559; Pc = 2.76 × 10-5, OR = 0.589, respectively). PRKCD ATG and CARD9 GCTTA haplotypes decreased susceptibility to VKH (Pc = 3.11 × 10-3, OR = 0.594; Pc = 5.00 × 10-3, OR = 0.639, respectively). Functional studies on rs3812555 genotyped individuals revealed that CC carriers had significantly higher CARD9 mRNA expression and tumour necrosis factor-α production than TC/TT carriers (P = 1.00 × 10-4; P = 2.00 × 10-3, respectively). CONCLUSIONS We found an association between PRKCD rs74437127 and CARD9 rs3812555 polymorphisms and VKH susceptibility and revealed that the increased susceptibility of rs3812555 for VKH may be mediated by regulating CARD9 gene expression and the production of pro-inflammatory cytokines, such as TNF-α.
Collapse
Affiliation(s)
- Chunya Zhou
- grid.488542.70000 0004 1758 0435Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000 People’s Republic of China ,grid.256112.30000 0004 1797 9307Department of Ophthalmology and Optometry, The School of Medical Technology and Engineering, Fujian Medical University, Jiaotong Road 88, Fuzhou, 350004 People’s Republic of China
| | - Shiya Cai
- grid.488542.70000 0004 1758 0435Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000 People’s Republic of China ,grid.256112.30000 0004 1797 9307Department of Ophthalmology and Optometry, The School of Medical Technology and Engineering, Fujian Medical University, Jiaotong Road 88, Fuzhou, 350004 People’s Republic of China
| | - Yuhong Xie
- grid.488542.70000 0004 1758 0435Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000 People’s Republic of China ,grid.256112.30000 0004 1797 9307Department of Ophthalmology and Optometry, The School of Medical Technology and Engineering, Fujian Medical University, Jiaotong Road 88, Fuzhou, 350004 People’s Republic of China
| | - Zhen Zeng
- grid.488542.70000 0004 1758 0435Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000 People’s Republic of China ,grid.256112.30000 0004 1797 9307Department of Ophthalmology and Optometry, The School of Medical Technology and Engineering, Fujian Medical University, Jiaotong Road 88, Fuzhou, 350004 People’s Republic of China
| | - Jun Zhang
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016 China
| | - Guannan Su
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016 China
| | - Qiuying Wu
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016 China
| | - Xingsheng Ye
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016 China
| | - Qingfeng Cao
- grid.452206.70000 0004 1758 417XThe First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016 China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Youyi Road 1, Chongqing, 400016, China.
| | - Jianmin Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Fujian Medical University, Engineering Research Center of Assistive Technology for Visual Impairment, Fujian Province University, Quanzhou, 362000, People's Republic of China. .,Department of Ophthalmology and Optometry, The School of Medical Technology and Engineering, Fujian Medical University, Jiaotong Road 88, Fuzhou, 350004, People's Republic of China.
| |
Collapse
|
4
|
Getiye Y, Rice TA, Phillips BD, Carrillo DF, He G. Dysregulated lipolysis and lipophagy in lipid droplets of macrophages from high fat diet-fed obese mice. J Cell Mol Med 2022; 26:4825-4836. [PMID: 35962606 PMCID: PMC9465182 DOI: 10.1111/jcmm.17513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 07/23/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is associated with lipid droplet (LD) accumulation, dysregulated lipolysis and chronic inflammation. Previously, the caspase recruitment domain‐containing protein 9 (CARD9) has been identified as a potential contributor to obesity‐associated abnormalities including cardiac dysfunction. In the current study, we explored a positive feedback signalling cycle of dysregulated lipolysis, CARD9‐associated inflammation, impaired lipophagy and excessive LD accumulation in sustaining the chronic inflammation associated with obesity. C57BL/6 WT and CARD9−/− mice were fed with normal diet (ND, 12% fat) or a high fat diet (HFD, 45% fat) for 5 months. Staining of LDs from peritoneal macrophages (PMs) revealed a significant increase in the number of cells with LD and the number of LD per cell in the HFD‐fed WT but not CARD9−/− obese mice. Rather, CARD9 KO significantly increased the mean LD size. WT obese mice showed down regulation of lipolytic proteins with increased diacylglycerol (DAG) content, and CARD9 KO normalized DAG with restored lipolytic protein expression. The build‐up of DAG in the WT obese mice is further associated with activation of PKCδ, NF‐κB and p38 MAPK inflammatory signalling in a CARDD9‐dependent manner. Inhibition of adipose triglyceride lipase (ATGL) by Atglistatin (Atg) resulted in similar effects as in CARD9−/− mice. Interestingly, CARD9 KO and Atg treatment enhanced lipophagy. In conclusion, HFD feeding likely initiated a positive feedback signalling loop from dysregulated lipolysis, CARD9‐dependent inflammation, impaired lipophagy, to excessive LD accumulation and sustained inflammation. CARD9 KO and Atg treatment protected against the chronic inflammation by interrupting this feedforward cycle.
Collapse
Affiliation(s)
- Yohannes Getiye
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, Wyoming, USA
| | - Tatiana Angel Rice
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, Wyoming, USA
| | - Brandon D Phillips
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, Wyoming, USA
| | - Daniel Fidel Carrillo
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, Wyoming, USA
| | - Guanglong He
- School of Pharmacy, College of Health Sciences, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
5
|
Liu X, Jiang B, Hao H, Liu Z. CARD9 Signaling, Inflammation, and Diseases. Front Immunol 2022; 13:880879. [PMID: 35432375 PMCID: PMC9005907 DOI: 10.3389/fimmu.2022.880879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022] Open
Abstract
Caspase-recruitment domain 9 (CARD9) protein is expressed in many cells especially in immune cells, and is critically involved in the function of the innate and adaptive immune systems through extensive interactions between CARD9 and other signaling molecules including NF-κB and MAPK. CARD9-mediated signaling plays a central role in regulating inflammatory responses and oxidative stress through the productions of important cytokines and chemokines. Abnormalities of CARD9 and CARD9 signaling or CARD9 mutations or polymorphism are associated with a variety of pathological conditions including infections, inflammation, and autoimmune disorders. This review focuses on the function of CARD9 and CARD9-mediated signaling pathways, as well as interactions with other important signaling molecules in different cell types and the relations to specific disease conditions including inflammatory diseases, infections, tumorigenesis, and cardiovascular pathologies.
Collapse
Affiliation(s)
- Xuanyou Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Bimei Jiang
- Department of Pathophysiology, Central South University, Changsha, China
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
6
|
Cramer J. Medicinal chemistry of the myeloid C-type lectin receptors Mincle, Langerin, and DC-SIGN. RSC Med Chem 2021; 12:1985-2000. [PMID: 35024612 PMCID: PMC8672822 DOI: 10.1039/d1md00238d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
In their role as pattern-recognition receptors on cells of the innate immune system, myeloid C-type lectin receptors (CLRs) assume important biological functions related to immunity, homeostasis, and cancer. As such, this family of receptors represents an appealing target for therapeutic interventions for modulating the outcome of many pathological processes, in particular related to infectious diseases. This review summarizes the current state of research into glycomimetic or drug-like small molecule ligands for the CLRs Mincle, Langerin, and DC-SIGN, which have potential therapeutic applications in vaccine research and anti-infective therapy.
Collapse
Affiliation(s)
- Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University of Düsseldorf Universitätsstr. 1 40225 Düsseldorf Germany
| |
Collapse
|
7
|
|
8
|
Zajta E, Csonka K, Tóth A, Tiszlavicz L, Németh T, Orosz A, Novák Á, Csikós M, Vágvölgyi C, Mócsai A, Gácser A. Signaling through Syk or CARD9 Mediates Species-Specific Anti- Candida Protection in Bone Marrow Chimeric Mice. mBio 2021; 12:e0160821. [PMID: 34465030 PMCID: PMC8406149 DOI: 10.1128/mbio.01608-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 01/12/2023] Open
Abstract
The spleen tyrosine kinase (Syk) and the downstream adaptor protein CARD9 are crucial signaling molecules in antimicrobial immunity. Candida parapsilosis is an emerging fungal pathogen with a high incidence in neonates, while Candida albicans is the most common agent of candidiasis. While signaling through Syk/CARD9 promotes protective host mechanisms in response to C. albicans, its function in immunity against C. parapsilosis remains unclear. Here, we generated Syk-/- and CARD9-/- bone marrow chimeric mice to study the role of Syk/CARD9 signaling in immune responses to C. parapsilosis compared to C. albicans. We demonstrate various functions of this pathway (e.g., phagocytosis, phagosome acidification, and killing) in Candida-challenged, bone marrow-derived macrophages with differential involvement of Syk and CARD9 along with species-specific differences in cytokine production. We report that Syk-/- or CARD9-/- chimeras rapidly display high susceptibility to C. albicans, while C. parapsilosis infection exacerbates over a prolonged period in these animals. Thus, our results establish that Syk and CARD9 contribute to systemic resistance to C. parapsilosis and C. albicans differently. Additionally, we confirm prior studies but also detail new insights into the fundamental roles of both proteins in immunity against C. albicans. Our data further suggest that Syk has a more prominent influence on anti-Candida immunity than CARD9. Therefore, this study reinforces the Syk/CARD9 pathway as a potential target for anti-Candida immune therapy. IMPORTANCE While C. albicans remains the most clinically significant Candida species, C. parapsilosis is an emerging pathogen with increased affinity to neonates. Syk/CARD9 signaling is crucial in immunity to C. albicans, but its role in in vivo responses to other pathogenic Candida species is largely unexplored. We used mice with hematopoietic systems deficient in Syk or CARD9 to comparatively study the function of these proteins in anti-Candida immunity. We demonstrate that Syk/CARD9 signaling has a protective role against C. parapsilosis differently than against C. albicans. Thus, this study is the first to reveal that Syk can exert immune responses during systemic Candida infections species specifically. Additionally, Syk-dependent immunity to a nonalbicans Candida species in an in vivo murine model has not been reported previously. We highlight that the contribution of Syk and CARD9 to fungal infections are not identical and underline this pathway as a promising immune-therapeutic target to fight Candida infections.
Collapse
Affiliation(s)
- Erik Zajta
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Katalin Csonka
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Adél Tóth
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | | | - Tamás Németh
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Anita Orosz
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Ádám Novák
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Máté Csikós
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Vágvölgyi
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Attila Gácser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- MTA-SZTE “Lendület” Mycobiome Research Group, University of Szeged, Szeged, Hungary
| |
Collapse
|
9
|
Reijneveld JF, Holzheimer M, Young DC, Lopez K, Suliman S, Jimenez J, Calderon R, Lecca L, Murray MB, Ishikawa E, Yamasaki S, Minnaard AJ, Moody DB, Van Rhijn I. Synthetic mycobacterial diacyl trehaloses reveal differential recognition by human T cell receptors and the C-type lectin Mincle. Sci Rep 2021; 11:2010. [PMID: 33479373 PMCID: PMC7820438 DOI: 10.1038/s41598-021-81474-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/04/2021] [Indexed: 11/10/2022] Open
Abstract
The cell wall of Mycobacterium tuberculosis is composed of diverse glycolipids which potentially interact with the human immune system. To overcome difficulties in obtaining pure compounds from bacterial extracts, we recently synthesized three forms of mycobacterial diacyltrehalose (DAT) that differ in their fatty acid composition, DAT1, DAT2, and DAT3. To study the potential recognition of DATs by human T cells, we treated the lipid-binding antigen presenting molecule CD1b with synthetic DATs and looked for T cells that bound the complex. DAT1- and DAT2-treated CD1b tetramers were recognized by T cells, but DAT3-treated CD1b tetramers were not. A T cell line derived using CD1b-DAT2 tetramers showed that there is no cross-reactivity between DATs in an IFN-γ release assay, suggesting that the chemical structure of the fatty acid at the 3-position determines recognition by T cells. In contrast with the lack of recognition of DAT3 by human T cells, DAT3, but not DAT1 or DAT2, activates Mincle. Thus, we show that the mycobacterial lipid DAT can be both an antigen for T cells and an agonist for the innate Mincle receptor, and that small chemical differences determine recognition by different parts of the immune system.
Collapse
Affiliation(s)
- Josephine F Reijneveld
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - Mira Holzheimer
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - David C Young
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Kattya Lopez
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA.,Socios En Salud, Lima, Peru
| | - Sara Suliman
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | | | | | | | - Megan B Murray
- Division of Global Health Equity, Department of Global Health and Social Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, Groningen, The Netherlands
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Ildiko Van Rhijn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Hale Building for Transformative Medicine, 60 Fenwood Road, Boston, MA, 02115, USA. .,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
10
|
Lindenwald DL, Lepenies B. C-Type Lectins in Veterinary Species: Recent Advancements and Applications. Int J Mol Sci 2020; 21:ijms21145122. [PMID: 32698416 PMCID: PMC7403975 DOI: 10.3390/ijms21145122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
C-type lectins (CTLs), a superfamily of glycan-binding receptors, play a pivotal role in the host defense against pathogens and the maintenance of immune homeostasis of higher animals and humans. CTLs in innate immunity serve as pattern recognition receptors and often bind to glycan structures in damage- and pathogen-associated molecular patterns. While CTLs are found throughout the whole animal kingdom, their ligand specificities and downstream signaling have mainly been studied in humans and in model organisms such as mice. In this review, recent advancements in CTL research in veterinary species as well as potential applications of CTL targeting in veterinary medicine are outlined.
Collapse
|
11
|
Mnich ME, van Dalen R, van Sorge NM. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front Cell Infect Microbiol 2020; 10:309. [PMID: 32733813 PMCID: PMC7358460 DOI: 10.3389/fcimb.2020.00309] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antigen-presenting cells (APCs) are present throughout the human body—in tissues, at barrier sites and in the circulation. They are critical for processing external signals to instruct both local and systemic responses toward immune tolerance or immune defense. APCs express an extensive repertoire of pattern-recognition receptors (PRRs) to detect and transduce these signals. C-type lectin receptors (CLRs) comprise a subfamily of PRRs dedicated to sensing glycans, including those expressed by commensal and pathogenic bacteria. This review summarizes recent findings on the recognition of and responses to bacteria by membrane-expressed CLRs on different APC subsets, which are discussed according to the primary site of infection. Many CLR-bacterial interactions promote bacterial clearance, whereas other interactions are exploited by bacteria to enhance their pathogenic potential. The discrimination between protective and virulence-enhancing interactions is essential to understand which interactions to target with new prophylactic or treatment strategies. CLRs are also densely concentrated at APC dendrites that sample the environment across intact barrier sites. This suggests an–as yet–underappreciated role for CLR-mediated recognition of microbiota-produced glycans in maintaining tolerance at barrier sites. In addition to providing a concise overview of identified CLR-bacteria interactions, we discuss the main challenges and potential solutions for the identification of new CLR-bacterial interactions, including those with commensal bacteria, and for in-depth structure-function studies on CLR-bacterial glycan interactions. Finally, we highlight the necessity for more relevant tissue-specific in vitro, in vivo and ex vivo models to develop therapeutic applications in this area.
Collapse
Affiliation(s)
- Malgorzata E Mnich
- Medical Microbiology, UMC Utrecht, Utrecht University, Utrecht, Netherlands.,GSK, Siena, Italy
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
12
|
Plant lectins and their usage in preparing targeted nanovaccines for cancer immunotherapy. Semin Cancer Biol 2020; 80:87-106. [PMID: 32068087 DOI: 10.1016/j.semcancer.2020.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/30/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Plant lectins, a natural source of glycans with a therapeutic potential may lead to the discovery of new targeted therapies. Glycans extracted from plant lectins are known to act as ligands for C-type lectin receptors (CLRs) that are primarily present on immune cells. Plant-derived glycosylated lectins offer diversity in their N-linked oligosaccharide structures that can serve as a unique source of homogenous and heterogenous glycans. Among the plant lectins-derived glycan motifs, Man9GlcNAc2Asn exhibits high-affinity interactions with CLRs that may resemble glycan motifs of pathogens. Thus, such glycan domains when presented along with antigens complexed with a nanocarrier of choice may bewilder the immune cells and direct antigen cross-presentation - a cytotoxic T lymphocyte immune response mediated by CD8+ T cells. Glycan structure analysis has attracted considerable interest as glycans are looked upon as better therapeutic alternatives than monoclonal antibodies due to their cost-effectiveness, reduced toxicity and side effects, and high specificity. Furthermore, this approach will be useful to understand whether the multivalent glycan presentation on the surface of nanocarriers can overcome the low-affinity lectin-ligand interaction and thereby modulation of CLR-dependent immune response. Besides this, understanding how the heterogeneity of glycan structure impacts the antigen cross-presentation is pivotal to develop alternative targeted therapies. In the present review, we discuss the findings on structural analysis of glycans from natural lectins performed using GlycanBuilder2 - a software tool based on a thorough literature review of natural lectins. Additionally, we discuss how multiple parameters like the orientation of glycan ligands, ligand density, simultaneous targeting of multiple CLRs and design of antigen delivery nanocarriers may influence the CLR targeting efficacy. Integrating this information will eventually set the ground for new generation immunotherapeutic vaccine design for the treatment of various human malignancies.
Collapse
|
13
|
Abstract
Chemical synthesis of trehalose glycolipids such as DAT, TDM, SL-1, SL-3, and Ac2SGL from MTb, emmyguyacins from fungi, succinoyl trehalose from rhodococcus, and maradolipids from worms, as well as mycobacterial oligosaccharides is reviewed.
Collapse
Affiliation(s)
- Santanu Jana
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai
- India
| | | |
Collapse
|
14
|
Alzahrani KS, Nadeem A, Ahmad SF, Al-Harbi NO, Ibrahim KE, El-Sherbeeny AM, Alhoshani AR, Alshammari MA, Alotaibi MR, Al-Harbi MM. Inhibition of spleen tyrosine kinase attenuates psoriasis-like inflammation in mice through blockade of dendritic cell-Th17 inflammation axis. Biomed Pharmacother 2018; 111:347-358. [PMID: 30593001 DOI: 10.1016/j.biopha.2018.12.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Psoriasis is a debilitating autoimmune disease of the skin characterized by acanthosis and hyperkeratosis resulting from excessive growth of keratinocytes in the epidermis and inflammatory infiltrates in the dermis. Innate immune cells such as dendritic cells (DCs), perform a critical role in the pathophysiology of psoriasis by presenting inflammatory/costimulatory signals for differentiation of Th17 cells. Recent studies point to the involvement of spleen tyrosine kinase (SYK) in inflammatory signaling cascade of DCs. However, it is yet to be determined whether SYK inhibition in DCs would lead to diminishment of psoriatic inflammation. Therefore, our study evaluated the effects of SYK inhibitor, R406 on imiquimod (IMQ)-induced psoriasis-like inflammation, expression of costimulatory/inflammatory molecules in DCs and their relationship with Th17/Treg cells. Our data show that R406 causes attenuation of IMQ-induced dermal inflammation as shown by reduction in ear/back skin thickness, acanthosis and myeloperoxidase activity. This was concurrent with reduction in inflammatory cytokines and co-stimulatory molecules in CD11c + DCs such as IL-6, IL-23, MHCII, and CD40. This favoured the suppression of Th17 cells and upregulation of Treg cells in R406-treated mice with psoriasis-like inflammation. Direct activation of TLR7 by IMQ in splenocytic cultures led to increased SYK expression in CD11c + DCs and release of IL-23/IL-6. IMQ-induced IL-6/IL-23 levels were significantly diminished by SYK inhibitor, R406 in splenocytic cultures. In essence, our study shows that SYK inhibition supresses psoriasis-like inflammation by modifying DC function in mice. Further, it implies that SYK inhibition could be a prospective therapeutic approach for the treatment of psoriasis-like inflammation.
Collapse
Affiliation(s)
- Khalid S Alzahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad M El-Sherbeeny
- Industrial Engineering Department, College of Engineering, King Saud University, Riyadh, Saudi Arabia
| | - Ali R Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed M Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Decout A, Silva-Gomes S, Drocourt D, Blattes E, Rivière M, Prandi J, Larrouy-Maumus G, Caminade AM, Hamasur B, Källenius G, Kaur D, Dobos KM, Lucas M, Sutcliffe IC, Besra GS, Appelmelk BJ, Gilleron M, Jackson M, Vercellone A, Tiraby G, Nigou J. Deciphering the molecular basis of mycobacteria and lipoglycan recognition by the C-type lectin Dectin-2. Sci Rep 2018; 8:16840. [PMID: 30443026 PMCID: PMC6237770 DOI: 10.1038/s41598-018-35393-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/05/2018] [Indexed: 01/04/2023] Open
Abstract
Dectin-2 is a C-type lectin involved in the recognition of several pathogens such as Aspergillus fumigatus, Candida albicans, Schistosoma mansonii, and Mycobacterium tuberculosis that triggers Th17 immune responses. Identifying pathogen ligands and understanding the molecular basis of their recognition is one of the current challenges. Purified M. tuberculosis mannose-capped lipoarabinomannan (ManLAM) was shown to induce signaling via Dectin-2, an activity that requires the (α1 → 2)-linked mannosides forming the caps. Here, using isogenic M. tuberculosis mutant strains, we demonstrate that ManLAM is a bona fide and actually the sole ligand mediating bacilli recognition by Dectin-2, although M. tuberculosis produces a variety of cell envelope mannoconjugates, such as phosphatidyl-myo-inositol hexamannosides, lipomannan or manno(lipo)proteins, that bear (α1 → 2)-linked mannosides. In addition, we found that Dectin-2 can recognize lipoglycans from other bacterial species, such as Saccharotrix aerocolonigenes or the human opportunistic pathogen Tsukamurella paurometabola, suggesting that lipoglycans are prototypical Dectin-2 ligands. Finally, from a structure/function relationship perspective, we show, using lipoglycan variants and synthetic mannodendrimers, that dimannoside caps and multivalent interaction are required for ligand binding to and signaling via Dectin-2. Better understanding of the molecular basis of ligand recognition by Dectin-2 will pave the way for the rational design of potent adjuvants targeting this receptor.
Collapse
Affiliation(s)
- Alexiane Decout
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France.,InvivoGen, Research Department, 31400, Toulouse, France.,Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Sandro Silva-Gomes
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France.,GlaxoSmithKline (GSK), Stevenage Herts, SG1 2NY, UK
| | | | - Emilyne Blattes
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France.,Innovative Medecine for Tuberculosis (iM4TB), Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Michel Rivière
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France
| | - Jacques Prandi
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France
| | - Gérald Larrouy-Maumus
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France.,Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France
| | - Beston Hamasur
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.,Biopromic AB, 171 65, Solna, Sweden
| | - Gunilla Källenius
- Department of Medicine, Karolinska Institutet Solna 171 76, Stockholm, Sweden
| | - Devinder Kaur
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA.,Massachusetts Supranational TB Reference Laboratory, University of Massachusetts Medical School, Jamaica Plain, MA, 0213, USA
| | - Karen M Dobos
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA
| | - Megan Lucas
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA
| | - Iain C Sutcliffe
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - Gurdyal S Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ben J Appelmelk
- Department of Medical Microbiology and Infection Control, VU University Medical Center, 1081 BT, Amsterdam, The Netherlands
| | - Martine Gilleron
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523-1682, USA
| | - Alain Vercellone
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France
| | - Gérard Tiraby
- InvivoGen, Research Department, 31400, Toulouse, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Université Paul Sabatier, 31077, Toulouse, France.
| |
Collapse
|
16
|
Bird JH, Khan AA, Nishimura N, Yamasaki S, Timmer MSM, Stocker BL. Synthesis of Branched Trehalose Glycolipids and Their Mincle Agonist Activity. J Org Chem 2018; 83:7593-7605. [DOI: 10.1021/acs.joc.7b03269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jessie H. Bird
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Ashna A. Khan
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Naoya Nishimura
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Sho Yamasaki
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| |
Collapse
|
17
|
Chiffoleau E. C-Type Lectin-Like Receptors As Emerging Orchestrators of Sterile Inflammation Represent Potential Therapeutic Targets. Front Immunol 2018; 9:227. [PMID: 29497419 PMCID: PMC5818397 DOI: 10.3389/fimmu.2018.00227] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/26/2018] [Indexed: 01/19/2023] Open
Abstract
Over the last decade, C-type lectin-like receptors (CTLRs), expressed mostly by myeloid cells, have gained increasing attention for their role in the fine tuning of both innate and adaptive immunity. Not only CTLRs recognize pathogen-derived ligands to protect against infection but also endogenous ligands such as self-carbohydrates, proteins, or lipids to control homeostasis and tissue injury. Interestingly, CTLRs act as antigen-uptake receptors via their carbohydrate-recognition domain for internalization and subsequent presentation to T-cells. Furthermore, CTLRs signal through a complex intracellular network leading to the secretion of a particular set of cytokines that differently polarizes downstream effector T-cell responses according to the ligand and pattern recognition receptor co-engagement. Thus, by orchestrating the balance between inflammatory and resolution pathways, CTLRs are now considered as driving players of sterile inflammation whose dysregulation leads to the development of various pathologies such as autoimmune diseases, allergy, or cancer. For examples, the macrophage-inducible C-type lectin (MINCLE), by sensing glycolipids released during cell-damage, promotes skin allergy and the pathogenesis of experimental autoimmune uveoretinitis. Besides, recent studies described that tumors use physiological process of the CTLRs’ dendritic cell-associated C-type lectin-1 (DECTIN-1) and MINCLE to locally suppress myeloid cell activation and promote immune evasion. Therefore, we aim here to overview the current knowledge of the pivotal role of CTLRs in sterile inflammation with special attention given to the “Dectin-1” and “Dectin-2” families. Moreover, we will discuss the potential of these receptors as promising therapeutic targets to treat a wide range of acute and chronic diseases.
Collapse
Affiliation(s)
- Elise Chiffoleau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,IHU Cesti, Nantes, France.,Labex Immunotherapy Graft Oncology (IGO), Nantes, France
| |
Collapse
|
18
|
Braganza CD, Teunissen T, Timmer MSM, Stocker BL. Identification and Biological Activity of Synthetic Macrophage Inducible C-Type Lectin Ligands. Front Immunol 2018; 8:1940. [PMID: 29387054 PMCID: PMC5776103 DOI: 10.3389/fimmu.2017.01940] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/15/2017] [Indexed: 11/15/2022] Open
Abstract
The macrophage inducible C-type lectin (Mincle) is a pattern recognition receptor able to recognize both damage-associated and pathogen-associated molecular patterns, and in this respect, there has been much interest in determining the scope of ligands that bind Mincle and how structural modifications to these ligands influence ensuing immune responses. In this review, we will present Mincle ligands of known chemical structure, with a focus on ligands that have been synthetically prepared, such as trehalose glycolipids, glycerol-based ligands, and 6-acylated glucose and mannose derivatives. The ability of the different classes of ligands to influence the innate, and consequently, the adaptive, immune response will be described, and where appropriate, structure-activity relationships within each class of Mincle ligands will be presented.
Collapse
Affiliation(s)
- Chriselle D. Braganza
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Thomas Teunissen
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
19
|
Ostrop J, Lang R. Contact, Collaboration, and Conflict: Signal Integration of Syk-Coupled C-Type Lectin Receptors. THE JOURNAL OF IMMUNOLOGY 2017; 198:1403-1414. [PMID: 28167651 DOI: 10.4049/jimmunol.1601665] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Several spleen tyrosine kinase-coupled C-type lectin receptors (CLRs) have emerged as important pattern recognition receptors for infectious danger. Because encounter with microbial pathogens leads to the simultaneous ligation of several CLRs and TLRs, the signals emanating from different pattern recognition receptors have to be integrated to achieve appropriate biological responses. In this review, we briefly summarize current knowledge about ligand recognition and core signaling by Syk-coupled CLRs. We then address mechanisms of synergistic and antagonistic crosstalk between different CLRs and with TLRs. Emerging evidence suggests that signal integration occurs through 1) direct interaction between receptors, 2) regulation of expression levels and localization, and 3) collaborative or conflicting signaling interference. Accordingly, we aim to provide a conceptual framework for the complex and sometimes unexpected outcome of CLR ligation in bacterial and fungal infection.
Collapse
Affiliation(s)
- Jenny Ostrop
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; .,Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; and
| | - Roland Lang
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
20
|
Patin EC, Orr SJ, Schaible UE. Macrophage Inducible C-Type Lectin As a Multifunctional Player in Immunity. Front Immunol 2017; 8:861. [PMID: 28791019 PMCID: PMC5525440 DOI: 10.3389/fimmu.2017.00861] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/07/2017] [Indexed: 01/08/2023] Open
Abstract
The macrophage-inducible C-type lectin (Mincle) is an innate immune receptor on myeloid cells sensing diverse entities including pathogens and damaged cells. Mincle was first described as a receptor for the mycobacterial cell wall glycolipid, trehalose-6,6′-dimycolate, or cord factor, and the mammalian necrotic cell-derived alarmin histone deacetylase complex unit Sin3-associated protein 130. Upon engagement by its ligands, Mincle induces secretion of innate cytokines and other immune mediators modulating inflammation and immunity. Since its discovery more than 25 years ago, the understanding of Mincle’s immune function has made significant advances in recent years. In addition to mediating immune responses to infectious agents, Mincle has been linked to promote tumor progression, autoimmunity, and sterile inflammation; however, further studies are required to completely unravel the complex role of Mincle in these distinct host responses. In this review, we discuss recent findings on Mincle’s biology with an emphasis on its diverse functions in immunity.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Priority Area Infections, Department Cellular Microbiology, Forschungszentrum Borstel, and German Center for Infection Research, TTU-TB, Borstel, Germany.,Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Selinda Jane Orr
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ulrich E Schaible
- Priority Area Infections, Department Cellular Microbiology, Forschungszentrum Borstel, and German Center for Infection Research, TTU-TB, Borstel, Germany
| |
Collapse
|
21
|
Abstract
The advances in subunit vaccines development have intensified the search for potent adjuvants, particularly adjuvants inducing cell-mediated immune responses. Identification of the C-type lectin Mincle as one of the receptors underlying the remarkable immunogenicity of the mycobacterial cell wall, via recognition of trehalose-6,6'-dimycolate (TDM), has opened avenues for the rational design of such molecules. Using a combination of chemical synthesis, biological evaluation, molecular dynamics simulations, and protein mutagenesis, we gained insight into the molecular bases of glycolipid recognition by Mincle. Unexpectedly, the fine structure of the fatty acids was found to play a key role in the binding of a glycolipid to the carbohydrate recognition domain of the lectin. Glucose and mannose esterified at O-6 by a synthetic α-ramified 32-carbon fatty acid showed agonist activity similar to that of TDM, despite their much simpler structure. Moreover, they were seen to stimulate proinflammatory cytokine production in primary human and murine cells in a Mincle-dependent fashion. Finally, they were found to induce strong Th1 and Th17 immune responses in vivo in immunization experiments in mice and conferred protection in a murine model of Mycobacterium tuberculosis infection. Here we describe the rational development of new molecules with powerful adjuvant properties.
Collapse
|
22
|
van Haren SD, Dowling DJ, Foppen W, Christensen D, Andersen P, Reed SG, Hershberg RM, Baden LR, Levy O. Age-Specific Adjuvant Synergy: Dual TLR7/8 and Mincle Activation of Human Newborn Dendritic Cells Enables Th1 Polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:4413-4424. [PMID: 27793997 PMCID: PMC7386828 DOI: 10.4049/jimmunol.1600282] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 09/26/2016] [Indexed: 02/07/2023]
Abstract
Due to functionally distinct cell-mediated immunity, newborns and infants are highly susceptible to infection with intracellular pathogens. Indeed, neonatal Ag-presenting dendritic cells (DCs) demonstrate impaired Th1 responses to many candidate adjuvants, including most TLR agonists (TLRAs). Combination adjuvantation systems may provide enhanced immune activation but have typically been developed without regard to the age of the target population. We posited that distinct combinations of TLRAs and C-type lectin receptor agonists may enhance Th1 responses of newborn DCs. TLRA/C-type lectin receptor agonist combinations were screened for enhancement of TNF production by human newborn and adult monocyte-derived DCs cultured in 10% autologous plasma or in newborn cord, infant, adult, and elderly whole blood. Monocyte-derived DC activation was characterized by targeted gene expression analysis, caspase-1 and NF-κB studies, cytokine multiplex and naive autologous CD4+ T cell activation. Dual activation of newborn DCs via the C-type lectin receptor, macrophage-inducible C-type lectin (trehalose-6,6-dibehenate), and TLR7/8 (R848) greatly enhanced caspase-1 and NF-κB activation, Th1 polarizing cytokine production and autologous Th1 polarization. Combined activation via TLR4 (glycopyranosyl lipid adjuvant aqueous formulation) and Dectin-1 (β-glucan peptide) acted synergistically in newborns and adults, but to a lesser extent. The degree of synergy varied dramatically with age, and was the greatest in newborns and infants with less synergy in adults and elders. Overall, combination adjuvant systems demonstrate markedly different immune activation with age, with combined DC activation via Macrophage-inducible C-type lectin and TLR7/8 representing a novel approach to enhance the efficacy of early-life vaccines.
Collapse
Affiliation(s)
- Simon D van Haren
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - David J Dowling
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Willemina Foppen
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, DK-2300, Denmark
| | - Peter Andersen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, DK-2300, Denmark
| | - Steven G Reed
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115
- Infectious Disease Research Institute, Seattle, WA 98102
| | | | - Lindsey R Baden
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115
- Division of Infectious Diseases, Brigham and Women's Hospital Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115
| | - Ofer Levy
- Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA 02115;
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| |
Collapse
|
23
|
Wilbers RHP, Westerhof LB, van de Velde J, Smant G, van Raaij DR, Sonnenberg ASM, Bakker J, Schots A. Physical Interaction of T Cells with Dendritic Cells Is Not Required for the Immunomodulatory Effects of the Edible Mushroom Agaricus subrufescens. Front Immunol 2016; 7:519. [PMID: 27920777 PMCID: PMC5118454 DOI: 10.3389/fimmu.2016.00519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/08/2016] [Indexed: 11/13/2022] Open
Abstract
Mushrooms are well known for their immunomodulating capacities. However, little is known about how mushroom-stimulated dendritic cells (DCs) affect T cells. Therefore, we investigated the effect of mushroom compounds derived from seven edible mushroom species on DCs, their fate in DCs, and the effect of the mushroom-stimulated DCs on T cells. Each mushroom species stimulated DCs in a different manner as was revealed from the DC’s cytokine response. Assessing DC maturation revealed that only one mushroom species, Agaricus subrufescens, induced complete DC maturation. The other six mushroom species upregulated MHC-II and CD86 expression, but did not significantly affect the expression of CD40 and CD11c. Nevertheless, mushroom compounds of all investigated mushroom species are endocytosed by DCs. Endocytosis is most likely mediated by C-type lectin receptors (CLRs) because CLR binding is Ca2+ dependent, and EGTA reduces TNF-α secretion with more than 90%. Laminarin partly inhibited TNF-α secretion indicating that the CLR dectin-1, among other CLRs, is involved in binding mushroom compounds. Stimulated DCs were shown to stimulate T cells; however, physical contact of DCs and T cells is not required. Because CLRs seem to play a prominent role in DC stimulation, mushrooms may function as a carbohydrate containing adjuvant to be used in conjunction with anti-fungal vaccines.
Collapse
Affiliation(s)
- Ruud H P Wilbers
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Lotte B Westerhof
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Jan van de Velde
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Geert Smant
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Debbie R van Raaij
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Anton S M Sonnenberg
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Jaap Bakker
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| | - Arjen Schots
- Plant Sciences Group, Wageningen University and Research Centre , Wageningen , Netherlands
| |
Collapse
|
24
|
Berzi A, Ordanini S, Joosten B, Trabattoni D, Cambi A, Bernardi A, Clerici M. Pseudo-Mannosylated DC-SIGN Ligands as Immunomodulants. Sci Rep 2016; 6:35373. [PMID: 27734954 PMCID: PMC5062166 DOI: 10.1038/srep35373] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
DC-SIGN, a C-type lectin mainly expressed by DCs, mediates antigen uptake and can induce specific immune responses, depending on the ligand involved. Owing to these properties, DC-SIGN is an attracting target for approaches aimed at tailoring the immune response towards specific immunologic outcomes. A multivalent DC-SIGN ligand (Polyman26), containing at its core a fluorescent "rod-like" spacer and able to inhibit DC-SIGN mediated HIV infection in nanomolar concentration, has been recently developed by our group. We investigated the internalization pattern and the ability of Polyman26 to elicit innate immune responses. Results obtained by confocal microscopy indicate that Polyman26 is internalized by DCs via receptor- mediated endocytosis and is then routed to endolysosomal compartments, thus being presented together with MHC class II molecules, with important implications for the development of vaccines. Moreover, Polyman26 up-regulated the production of β-chemokines and pro-inflammatory cytokines (including IL-1β, IL-6, IL-12, and TNFα) as well as the expression of TLR9 and CD40L. These results indicate that glycomimetic DC-SIGN ligands should be further investigated and suggest that these compounds could be used to differentially stimulate immune responses.
Collapse
Affiliation(s)
- Angela Berzi
- Chair of Immunology, Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Stefania Ordanini
- Department of Chemistry, University of Milan, Via C. Golgi 19, 20133, Milan, Italy
| | - Ben Joosten
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 9101 6500 HB, Nijmegen, The Netherlands
| | - Daria Trabattoni
- Chair of Immunology, Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Alessandra Cambi
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 9101 6500 HB, Nijmegen, The Netherlands
| | - Anna Bernardi
- Department of Chemistry, University of Milan, Via C. Golgi 19, 20133, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Via F.lli VCervi 93, 20090 Milan, Italy.,Don C. Gnocchi Foundation, IRCCS, Via Capecelatro 66, 20148 Milan, Italy
| |
Collapse
|
25
|
Cao L, Qin X, Peterson MR, Haller SE, Wilson KA, Hu N, Lin X, Nair S, Ren J, He G. CARD9 knockout ameliorates myocardial dysfunction associated with high fat diet-induced obesity. J Mol Cell Cardiol 2016; 92:185-95. [PMID: 26900039 PMCID: PMC4904726 DOI: 10.1016/j.yjmcc.2016.02.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/26/2016] [Accepted: 02/16/2016] [Indexed: 01/07/2023]
Abstract
Obesity is associated with chronic inflammation which plays a critical role in the development of cardiovascular dysfunction. Because the adaptor protein caspase recruitment domain-containing protein 9 (CARD9) in macrophages regulates innate immune responses via activation of pro-inflammatory cytokines, we hypothesize that CARD9 mediates the pro-inflammatory signaling associated with obesity en route to myocardial dysfunction. C57BL/6 wild-type (WT) and CARD9(-/-) mice were fed normal diet (ND, 12% fat) or a high fat diet (HFD, 45% fat) for 5months. At the end of 5-month HFD feeding, cardiac function was evaluated using echocardiography. Cardiomyocytes were isolated and contractile properties were measured. Immunofluorescence was performed to detect macrophage infiltration in the heart. Heart tissue homogenates, plasma, and supernatants from isolated macrophages were collected to measure the concentrations of pro-inflammatory cytokines using ELISA kits. Western immunoblotting analyses were performed on heart tissue homogenates and isolated macrophages to explore the underlying signaling mechanism(s). CARD9 knockout alleviated HFD-induced insulin resistance and glucose intolerance, prevented myocardial dysfunction with preserved cardiac fractional shortening and cardiomyocyte contractile properties. CARD9 knockout also significantly decreased the number of infiltrated macrophages in the heart with reduced myocardium-, plasma-, and macrophage-derived cytokines including IL-6, IL-1β and TNFα. Finally, CARD9 knockout abrogated the increase of p38 MAPK phosphorylation, the decrease of LC3BII/LC3BI ratio and the up-regulation of p62 expression in the heart induced by HFD feeding and restored cardiac autophagy signaling. In conclusion, CARD9 knockout ameliorates myocardial dysfunction associated with HFD-induced obesity, potentially through reduction of macrophage infiltration, suppression of p38 MAPK phosphorylation, and preservation of autophagy in the heart.
Collapse
Affiliation(s)
- Li Cao
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA; College of Pharmaceutical Sciences, Soochow University, Soochow, Jiangsu 215123, PR China
| | - Xing Qin
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Matthew R Peterson
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Samantha E Haller
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Kayla A Wilson
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Nan Hu
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Xin Lin
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sreejayan Nair
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Jun Ren
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA
| | - Guanglong He
- School of Pharmacy, University of Wyoming, College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
26
|
Zimmermann S, Lepenies B. Glycans as Vaccine Antigens and Adjuvants: Immunological Considerations. Methods Mol Biol 2015; 1331:11-26. [PMID: 26169732 DOI: 10.1007/978-1-4939-2874-3_2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carbohydrates can be found on the cell surface of nearly every cell ranging from bacteria to fungi right up to mammalian cells. Carbohydrates and their interactions with carbohydrate-binding proteins play crucial roles in multiple biological processes including immunity, homeostasis, cellular communication, cell migration, and the regulation of serum glycoprotein levels. In the last decades, the interest in exploiting the biological activity of glycans as vaccine components has considerably increased. On the one hand, carbohydrates display epitopes to generate protective antibodies against pathogen-derived cell wall structures and on the other hand, glycans have the potential to stimulate the immune system; thus they can act as potent vaccine adjuvants.An effective vaccine consists of two major components, the vaccine antigen and an adjuvant. The vaccine antigen is an original or modified part of the pathogen that causes the disease. The immune response triggered by vaccination should induce antigen-specific plasma cells secreting protective antibodies as well as the development of memory T and B cells. Carbohydrate structures on pathogens represent an important class of antigens that can activate B cells to produce protective anti-carbohydrate antibodies in adults. A major breakthrough in vaccine development was the design of conjugate vaccines that evoke protective antibody responses against encapsulated bacteria strains such as Haemophilus influenzae, Streptococcus pneumoniae, or Neisseria meningitidis in adults, but also in young children. The first part of this chapter focuses on immune responses triggered by carbohydrate-based vaccines. The second part of the chapter discusses the immunological mechanisms of carbohydrate-based adjuvants to increase the immunogenicity of vaccines.
Collapse
Affiliation(s)
- Stephanie Zimmermann
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476, Potsdam, Germany,
| | | |
Collapse
|
27
|
Vázquez A, Ruiz-Rosado JDD, Terrazas LI, Juárez I, Gomez-Garcia L, Calleja E, Camacho G, Chávez A, Romero M, Rodriguez T, Espinoza B, Rodriguez-Sosa M. Mouse macrophage galactose-type lectin (mMGL) is critical for host resistance against Trypanosoma cruzi infection. Int J Biol Sci 2014; 10:909-20. [PMID: 25170304 PMCID: PMC4147224 DOI: 10.7150/ijbs.9214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/05/2014] [Indexed: 12/27/2022] Open
Abstract
The C-type lectin receptor mMGL is expressed exclusively by myeloid antigen presenting cells (APC) such as dendritic cells (DC) and macrophages (Mφ), and it mediates binding to glycoproteins carrying terminal galactose and α- or β-N-acetylgalactosamine (Gal/GalNAc) residues. Trypanosoma cruzi (T. cruzi) expresses large amounts of mucin (TcMUC)-like glycoproteins. Here, we show by lectin-blot that galactose moieties are also expressed on the surface of T. cruzi. Male mMGL knockout (-/-) and wild-type (WT) C57BL/6 mice were infected intraperitoneally with 104T. cruzi trypomastigotes (Queretaro strain). Following T. cruzi infection, mMGL-/- mice developed higher parasitemia and higher mortality rates compared with WT mice. Although hearts from T. cruzi-infected WT mice presented few amastigote nests, mMGL-/- mice displayed higher numbers of amastigote nests. Compared with WT, Mφ from mMGL-/- mice had low production of nitric oxide (NO), interleukin (IL)-12 and tumor necrosis factor (TNF)-α in response to soluble T. cruzi antigens (TcAg). Interestingly, upon in vitro T. cruzi infection, mMGL-/- Mφ expressed lower levels of MHC-II and TLR-4 and harbored higher numbers of parasites, even when mMGL-/- Mφ were previously primed with IFN-γ or LPS/IFN-γ. These data suggest that mMGL plays an important role during T. cruzi infection, is required for optimal Mφ activation, and may synergize with TLR-4-induced pathways to produce TNF-α, IL-1β and NO during the early phase of infection.
Collapse
Affiliation(s)
- Alicia Vázquez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Juan de Dios Ruiz-Rosado
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Luis I Terrazas
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Imelda Juárez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Lorena Gomez-Garcia
- 2. Department of Immunology, Instituto Nacional de Cardiología "Ignacio Chávez," México, D.F. 14080 México
| | - Elsa Calleja
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Griselda Camacho
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Ana Chávez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Miriam Romero
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Tonathiu Rodriguez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Bertha Espinoza
- 3. Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México. México, D.F. 04510 México
| | - Miriam Rodriguez-Sosa
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| |
Collapse
|
28
|
Aretz J, Wamhoff EC, Hanske J, Heymann D, Rademacher C. Computational and experimental prediction of human C-type lectin receptor druggability. Front Immunol 2014; 5:323. [PMID: 25071783 PMCID: PMC4090677 DOI: 10.3389/fimmu.2014.00323] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/26/2014] [Indexed: 01/21/2023] Open
Abstract
Mammalian C-type lectin receptors (CTLRS) are involved in many aspects of immune cell regulation such as pathogen recognition, clearance of apoptotic bodies, and lymphocyte homing. Despite a great interest in modulating CTLR recognition of carbohydrates, the number of specific molecular probes is limited. To this end, we predicted the druggability of a panel of 22 CTLRs using DoGSiteScorer. The computed druggability scores of most structures were low, characterizing this family as either challenging or even undruggable. To further explore these findings, we employed a fluorine-based nuclear magnetic resonance screening of fragment mixtures against DC-SIGN, a receptor of pharmacological interest. To our surprise, we found many fragment hits associated with the carbohydrate recognition site (hit rate = 13.5%). A surface plasmon resonance-based follow-up assay confirmed 18 of these fragments (47%) and equilibrium dissociation constants were determined. Encouraged by these findings we expanded our experimental druggability prediction to Langerin and MCL and found medium to high hit rates as well, being 15.7 and 10.0%, respectively. Our results highlight limitations of current in silico approaches to druggability assessment, in particular, with regard to carbohydrate-binding proteins. In sum, our data indicate that small molecule ligands for a larger panel of CTLRs can be developed.
Collapse
Affiliation(s)
- Jonas Aretz
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany ; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin , Berlin , Germany
| | - Eike-Christian Wamhoff
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany ; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin , Berlin , Germany
| | - Jonas Hanske
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany ; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin , Berlin , Germany
| | - Dario Heymann
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany ; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin , Berlin , Germany
| |
Collapse
|
29
|
Fungal Engagement of the C-Type Lectin Mincle Suppresses Dectin-1-Induced Antifungal Immunity. Cell Host Microbe 2014; 15:494-505. [DOI: 10.1016/j.chom.2014.03.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 02/13/2014] [Accepted: 03/17/2014] [Indexed: 01/15/2023]
|
30
|
Berzi A, Varga N, Sattin S, Antonazzo P, Biasin M, Cetin I, Trabattoni D, Bernardi A, Clerici M. Pseudo-mannosylated DC-SIGN ligands as potential adjuvants for HIV vaccines. Viruses 2014; 6:391-403. [PMID: 24473338 PMCID: PMC3939462 DOI: 10.3390/v6020391] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/07/2014] [Accepted: 01/20/2014] [Indexed: 11/17/2022] Open
Abstract
The development of new and effective adjuvants may play a fundamental role in improving HIV vaccine efficacy. New classes of vaccine adjuvants activate innate immunity receptors, notably toll like receptors (TLRs). Adjuvants targeting the C-Type lectin receptor DC-SIGN may be alternative or complementary to adjuvants based on TRL activation. Herein we evaluate the ability of the glycomimetic DC-SIGN ligand Polyman 19 (PM 19) to modulate innate immune responses. Results showed that PM 19 alone, or in combination with TLR agonists, induces the expression of cytokines, β chemokines and co-stimulatory molecules that may, in turn, modulate adaptive immunity and exert anti-viral effects. These results indicate that the suitability of this compound as a vaccine adjuvant should be further evaluated.
Collapse
Affiliation(s)
- Angela Berzi
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via GB. Grassi 74, 20157 Milan, Italy.
| | - Norbert Varga
- Department of Chemistry, University of Milan, Via C.Golgi 19, 20133 Milan, Italy.
| | - Sara Sattin
- Department of Chemistry, University of Milan, Via C.Golgi 19, 20133 Milan, Italy.
| | - Patrizio Antonazzo
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via GB. Grassi 74, 20157 Milan, Italy.
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via GB. Grassi 74, 20157 Milan, Italy.
| | - Irene Cetin
- Unit of Obstetrics and Gynecology, Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via GB. Grassi 74, 20157 Milan, Italy.
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan, Via GB. Grassi 74, 20157 Milan, Italy.
| | - Anna Bernardi
- Department of Chemistry, University of Milan, Via C.Golgi 19, 20133 Milan, Italy.
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Via F.lli VCervi 93, 20090 Milan, Italy.
| |
Collapse
|
31
|
Burghardt KM, Avinashi V, Kosar C, Xu W, Wales PW, Avitzur Y, Muise A. A CARD9 polymorphism is associated with decreased likelihood of persistent conjugated hyperbilirubinemia in intestinal failure. PLoS One 2014; 9:e85915. [PMID: 24465786 PMCID: PMC3897546 DOI: 10.1371/journal.pone.0085915] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022] Open
Abstract
Recently, genetic associations have been described in intestinal transplants. Namely, Crohn's disease susceptibility gene NOD2 polymorphisms have been reported to be more prevalent in patients with graft failure following intestinal transplantation (IT). Therefore, we sought to determine if polymorphisms in the NOD2 signaling cascade, including NOD2, CARD9, RAC1 and ATG16L1 are associated with intestinal failure (IF) or its complications. We carried out a cross-sectional study of 59 children with IF and 500 healthy Caucasian controls. Using the Taqman platform we determined the prevalence of NOD2 as well as ATG16L1, RAC1 and CARD9 SNPs. NOD2 pathway polymorphisms were evaluated in relation to outcomes of episodes of sepsis, ICU admissions, hyperbilirubinemia and need for IT. We found that the minor allele of a CARD9 SNP was associated with protection from developing IF when compared to healthy controls and was also associated with decreased odds of sustained conjugated hyperbilirubinemia. Therefore, IF patients with CARD9 polymorphism are less likely to develop progressive liver disease and suggests that host innate immunity may play a role in IF associated liver disease.
Collapse
Affiliation(s)
- Karolina Maria Burghardt
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant Centre, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Toronto, Ontario, Canada
| | - Vishal Avinashi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, BC Children Hospital, Vancouver, British Columbia, Canada
| | - Christina Kosar
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant Centre, Toronto, Ontario, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Paul W. Wales
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant Centre, Toronto, Ontario, Canada
| | - Yaron Avitzur
- Group for Improvement of Intestinal Function and Treatment (GIFT), Transplant Centre, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Toronto, Ontario, Canada
- * E-mail: (YA); (AM)
| | - Aleixo Muise
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Toronto, Ontario, Canada
- SickKids IBD Centre, Program in Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (YA); (AM)
| |
Collapse
|
32
|
Stocker BL, Khan AA, Chee SH, Kamena F, Timmer MSM. On One Leg: Trehalose Monoesters Activate Macrophages in a Mincle-Dependent Manner. Chembiochem 2014; 15:382-8. [DOI: 10.1002/cbic.201300674] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Indexed: 11/10/2022]
|
33
|
Wang H, LeBert V, Hung CY, Galles K, Saijo S, Lin X, Cole GT, Klein BS, Wüthrich M. C-type lectin receptors differentially induce th17 cells and vaccine immunity to the endemic mycosis of North America. THE JOURNAL OF IMMUNOLOGY 2014; 192:1107-1119. [PMID: 24391211 DOI: 10.4049/jimmunol.1302314] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vaccine immunity to the endemic mycoses of North America requires Th17 cells, but the pattern recognition receptors and signaling pathways that drive these protective responses have not been defined. We show that C-type lectin receptors exert divergent contributions to the development of antifungal Th17 cells and vaccine resistance against Blastomyces dermatitidis, Histoplasma capsulatum, and Coccidioides posadasii. Acquired immunity to B. dermatitidis requires Dectin-2, whereas vaccination against H. capsulatum and C. posadasii infection depends on innate sensing by Dectin-1 and Dectin-2, but not Mincle. Tracking Ag-specific T cells in vivo established that the Card9 signaling pathway acts indispensably and exclusively on differentiation of Th17 cells, while leaving intact their activation, proliferation, survival, and migration. Whereas Card9 signaling is essential, C-type lectin receptors offer distinct and divergent contributions to vaccine immunity against these endemic fungal pathogens. Our work provides new insight into innate immune mechanisms that drive vaccine immunity and Th17 cells.
Collapse
Affiliation(s)
- Huafeng Wang
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792
| | - Vanessa LeBert
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792
| | - Chiung Yu Hung
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas
| | - Kevin Galles
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792
| | - Shinobu Saijo
- Center for Experimental Medicine and Systems Biology, University of Tokyo, Japan
| | - Xin Lin
- Department of Molecular and Cellular Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Garry T Cole
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, Texas
| | - Bruce S Klein
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792.,Department of Internal Medicine, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792.,Department of Medical Microbiology and Immunology, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792
| | - Marcel Wüthrich
- Department of Pediatrics, University of Wisconsin Medical School, University of Wisconsin Hospital and Clinics, Madison, WI, 53792
| |
Collapse
|
34
|
Feinberg H, Jégouzo SAF, Rowntree TJW, Guan Y, Brash MA, Taylor ME, Weis WI, Drickamer K. Mechanism for recognition of an unusual mycobacterial glycolipid by the macrophage receptor mincle. J Biol Chem 2013; 288:28457-65. [PMID: 23960080 PMCID: PMC3789947 DOI: 10.1074/jbc.m113.497149] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/16/2013] [Indexed: 12/31/2022] Open
Abstract
Binding of the macrophage lectin mincle to trehalose dimycolate, a key glycolipid virulence factor on the surface of Mycobacterium tuberculosis and Mycobacterium bovis, initiates responses that can lead both to toxicity and to protection of these pathogens from destruction. Crystallographic structural analysis, site-directed mutagenesis, and binding studies with glycolipid mimics have been used to define an extended binding site in the C-type carbohydrate recognition domain (CRD) of bovine mincle that encompasses both the headgroup and a portion of the attached acyl chains. One glucose residue of the trehalose Glcα1-1Glcα headgroup is liganded to a Ca(2+) in a manner common to many C-type CRDs, whereas the second glucose residue is accommodated in a novel secondary binding site. The additional contacts in the secondary site lead to a 36-fold higher affinity for trehalose compared with glucose. An adjacent hydrophobic groove, not seen in other C-type CRDs, provides a docking site for one of the acyl chains attached to the trehalose, which can be targeted with small molecule analogs of trehalose dimycolate that bind with 52-fold higher affinity than trehalose. The data demonstrate how mincle bridges between the surfaces of the macrophage and the mycobacterium and suggest the possibility of disrupting this interaction. In addition, the results may provide a basis for design of adjuvants that mimic the ability of mycobacteria to stimulate a response to immunization that can be employed in vaccine development.
Collapse
Affiliation(s)
- Hadar Feinberg
- From the Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305 and
| | - Sabine A. F. Jégouzo
- the Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | | | - Yue Guan
- the Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - Matthew A. Brash
- the Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - Maureen E. Taylor
- the Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - William I. Weis
- From the Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305 and
| | - Kurt Drickamer
- the Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| |
Collapse
|
35
|
Helbig ET, Opitz B, Sander LE. Adjuvant immunotherapies as a novel approach to bacterial infections. Immunotherapy 2013; 5:365-81. [PMID: 23557420 DOI: 10.2217/imt.13.17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The rapid emergence of multidrug-resistant pathogens, especially Gram-negative bacteria and mycobacteria, represents one of the major medical challenges of the 21st century. The gradual loss of effective classical antibiotics for many bacterial pathogens, combined with an increasing population density and mobility, urgently calls for the development of novel treatments. Here, we discuss the potential of adjuvant immunotherapies to selectively stimulate protective immune responses as a treatment option for bacterial infections. In order to elicit appropriate immune responses and to avoid unwanted inflammatory tissue damage, it is essential to identify ligands and receptor pathways that specifically control protective responses at the site of infection. We summarize existing data and discuss suitable candidate targets for future immunotherapies of infectious diseases.
Collapse
Affiliation(s)
- Elisa T Helbig
- Department of Infectious Diseases & Pulmonary Medicine, Charité University Hospital, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | |
Collapse
|
36
|
Lepenies B, Lee J, Sonkaria S. Targeting C-type lectin receptors with multivalent carbohydrate ligands. Adv Drug Deliv Rev 2013; 65:1271-81. [PMID: 23727341 DOI: 10.1016/j.addr.2013.05.007] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/19/2013] [Accepted: 05/22/2013] [Indexed: 01/08/2023]
Abstract
C-type lectin receptors (CLRs) represent a large receptor family including collectins, selectins, lymphocyte lectins, and proteoglycans. CLRs share a structurally homologous carbohydrate-recognition domain (CRD) and often bind carbohydrates in a Ca²⁺-dependent manner. In innate immunity, CLRs serve as pattern recognition receptors (PRRs) and bind to the glycan structures of pathogens and also to self-antigens. In nature, the low affinity of CLR/carbohydrate interactions is overcome by multivalent ligand presentation at the surface of cells or pathogens. Thus, multivalency is a promising strategy for targeting CLR-expressing cells and, indeed, carbohydrate-based targeting approaches have been employed for a number of CLRs, including asialoglycoprotein receptor (ASGPR) in the liver, or DC-SIGN expressed by dendritic cells. Since CLR engagement not only mediates endocytosis but also influences intracellular signaling pathways, CLR targeting may allow for cell-specific drug delivery and also the modulation of cellular functions. Glyconanoparticles, glycodendrimers, and glycoliposomes were successfully used as tools for CLR-specific targeting. This review will discuss different approaches for multivalent CLR ligand presentation and aims to highlight how CLR targeting has been employed for cell specific drug delivery. Major emphasis is directed towards targeting of CLRs expressed by antigen-presenting cells to modulate immune responses.
Collapse
|
37
|
Vukman KV, Ravidà A, Aldridge AM, O'Neill SM. Mannose receptor and macrophage galactose-type lectin are involved in Bordetella pertussis mast cell interaction. J Leukoc Biol 2013; 94:439-48. [PMID: 23794711 DOI: 10.1189/jlb.0313130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mast cells are crucial in the development of immunity against Bordetella pertussis, and the function of TLRs in this process has been investigated. Here, the interaction between mast cells and B. pertussis with an emphasis on the role of CLRs is examined. In this study, two CLRs, MGL and MR, were detected for the first time on the surface of mast cells. The involvement of MR and MGL in the stimulation of mast cells by heat-inactivated BP was investigated by the use of blocking antibodies and specific carbohydrate ligands. The cell wall LOS of BP was also isolated to explore its role in this interaction. Mast cells stimulated with heat-inactivated BP or BP LOS induced TNF-α, IL-6, and IFN-γ secretion, which was suppressed by blocking MR or MGL. Inhibition of CLRs signaling during BP stimulation affected the ability of mast cells to promote cytokine secretion in T cells but had no effect on the cell-surface expression of ICAM1. Blocking MR or MGL suppressed BP-induced NF-κB expression but not ERK phosphorylation. Syk was involved in the CLR-mediated activation of mast cells by BP. Bacterial recognition by immune cells has been predominantly attributed to TLRs; in this study, the novel role of CLRs in the BP-mast cell interaction is highlighted.
Collapse
Affiliation(s)
- Krisztina V Vukman
- Parasite Immune Modulation Group, School of Biotechnology, Faculty of Science and Health, Dublin City University, Glasnevin, Dublin, Ireland
| | | | | | | |
Collapse
|
38
|
Perrie Y, Kastner E, Kaur R, Wilkinson A, Ingham AJ. A case-study investigating the physicochemical characteristics that dictate the function of a liposomal adjuvant. Hum Vaccin Immunother 2013; 9:1374-81. [PMID: 23584249 DOI: 10.4161/hv.24694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A range of particulate delivery systems have been considered as vaccine adjuvants. Of these systems, liposomes offer a range of advantages including versatility and flexibility in design format and their ability to incorporate a range of immunomodulators and antigens. Here we briefly outline research, from within our laboratories, which focused on the systematic evaluation of cationic liposomes as vaccines adjuvants. Our aim was to identify physicochemical characteristics that correlate with vaccine efficacy, with particular consideration of the interlink between depot-forming action and immune responses. A variety of parameters were investigated and over a range of studies we have confirmed that cationic liposomes, based on dimethyldioctadecylammonium bromide and trehalose 6,6'-dibehenate formed a depot at the injection site, which stimulates recruitment of antigen presenting cells to the injection site and promotes strong humoral and cell-mediated immune responses. Physicochemical factors which promote a strong vaccine depot include the combination of a high cationic charge and electrostatic binding of the antigen to the liposome system and the use of lipids with high transition temperatures, which form rigid bilayer vesicles. Reduction in vesicle size of cationic vesicles did not promote enhanced drainage from the injection site. However, reducing the cationic nature through substitution of the cationic lipid for a neutral lipid, or by masking of the charge using PEGylation, resulted in a reduced depot formation and reduced Th1-type immune responses, while Th2-type responses were less influenced. These studies confirm that the physicochemical characteristics of particulate-based adjuvants play a key role in the modulation of immune responses.
Collapse
Affiliation(s)
- Yvonne Perrie
- School of Life and Health Sciences; Aston University; Birmingham, UK
| | | | | | | | | |
Collapse
|
39
|
Grimm SK, Ackerman ME. Vaccine design: emerging concepts and renewed optimism. Curr Opin Biotechnol 2013; 24:1078-88. [PMID: 23474232 DOI: 10.1016/j.copbio.2013.02.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 01/15/2023]
Abstract
Arguably, vaccination represents the single most effective medical intervention ever developed. Yet, vaccines have failed to provide any or adequate protection against some of the most significant global diseases. The pathogens responsible for these vaccine-recalcitrant diseases have properties that allow them to evade immune surveillance and misdirect or eliminate the immune response. However, genomic and systems biology tools, novel adjuvants and delivery systems, and refined molecular insight into protective immunity have started to redefine the landscape, and results from recent efficacy trials of HIV and malaria vaccines have instilled hope that another golden age of vaccines may be on the horizon.
Collapse
|
40
|
Abstract
We assessed the role of Dectin-1 in the immune response to the pathogenic fungus Coccidioides, both in vitro and in vivo, using mice with a targeted mutation in Clec7a. Elicited peritoneal macrophages responded to formalin-killed spherules (FKS) and alkali-treated FKS by secreting proinflammatory cytokines in a Dectin-1- and β-glucan-dependent manner. The responses of bone marrow-derived dendritic cells (BMDC) to the same stimulants were more complex; interleukin 1β (IL-1β) and tumor necrosis factor alpha (TNF-α) secretion was independent of Dectin-1, while IL-6, IL-10, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were largely but not entirely dependent on Dectin-1. After intranasal infection, Dectin-1(-/-) mice had lower concentrations of IL-12p70, gamma interferon (IFN-γ), IL-1β, and the Th17 cytokines IL-22, IL-23, and 17A in the lung lavage fluid, which may explain why they were significantly more susceptible to pulmonary coccidioidomycosis two weeks after infection. The Dectin-1 mutation was even more deleterious in (B6 × DBA/2)F(2) mice, which are more resistant to coccidioidomycosis than B6 mice by virtue of protective genes from DBA/2, a genetically resistant strain. We also found that two susceptible strains of mice (B6 and BALB/c) expressed much less Dectin-1 in their lungs than did resistant DBA/2 mice. We conclude that Dectin-1 is necessary for resistance to Coccidioides immitis, that Dectin-1 promotes both Th1 and Th17 protective immune responses to this infection, and that there is a correlation between expression of Dectin-1 by the inflammatory infiltrate and resistance to coccidioidomycosis. IMPORTANCE Coccidioidomycosis is a fungal infection endemic in the southwestern United States and neighboring Mexico, causing ~150,000 lung infections in people and resulting in ~17,000 hospitalizations annually in California alone. Very little is known about innate immunity to this fungus. This paper shows that Dectin-1, the primary β-glucan receptor on myeloid cells, is required for resistance to this pathogen. Dectin-1 is part of the innate immune system, and it is needed to direct the acquired immune response toward into a pathway that will lead to macrophage activation. Lungs from infected mice lacking Dectin-1 had lower concentrations of Th1 and Th17 cytokines, two cytokine pathways that are very important for acquired T cell immunity to Coccidioides spp. This is the first demonstration that Dectin-1 is required for host resistance to a dimorphic, primary pathogenic fungus.
Collapse
|
41
|
Zhu W, Ma H, Miao J, Huang G, Tong M, Zou S. β-Glucan modulates the lipopolysaccharide-induced innate immune response in rat mammary epithelial cells. Int Immunopharmacol 2013; 15:457-65. [DOI: 10.1016/j.intimp.2012.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 12/05/2012] [Accepted: 12/05/2012] [Indexed: 01/23/2023]
|
42
|
Lang R. Recognition of the mycobacterial cord factor by Mincle: relevance for granuloma formation and resistance to tuberculosis. Front Immunol 2013; 4:5. [PMID: 23355839 PMCID: PMC3553576 DOI: 10.3389/fimmu.2013.00005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/04/2013] [Indexed: 11/13/2022] Open
Abstract
The world's most successful intracellular bacterial pathogen, Mycobacterium tuberculosis (MTB), survives inside macrophages by blocking phagosome maturation and establishes chronic infection characterized by the formation of granulomas. Trehalose-6,6-dimycolate (TDM), the mycobacterial cord factor, is the most abundant cell wall lipid of virulent mycobacteria, is sufficient to cause granuloma formation, and has long been known to be a major virulence factor of MTB. Recently, TDM has been shown to activate the Syk-Card9 signaling pathway in macrophages through binding to the C-type lectin receptor Mincle. The Mincle-Card9 pathway is required for activation of macrophages by TDM in vitro and for granuloma formation in vivo following injection of TDM. Whether this pathway is also exploited by MTB to reprogram the macrophage into a comfortable niche has not been explored yet. Several recent studies have investigated the phenotype of Mincle-deficient mice in mycobacterial infection, yielding divergent results in terms of a role for Mincle in host resistance. Here, we review these studies, discuss possible reasons for discrepant results and highlight open questions in the role of Mincle and other C-type lectin receptors in the infection biology of MTB.
Collapse
Affiliation(s)
- Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg Erlangen, Germany
| |
Collapse
|
43
|
Abstract
Immunity to pathogens critically requires pattern-recognition receptors (PRRs) to trigger intracellular signaling cascades that initiate and direct innate and adaptive immune responses. For fungal infections, these responses are primarily mediated by members of the C-type lectin receptor family. In this Review, we highlight recent advances in the understanding of the roles and mechanisms of these multifunctional receptors, explore how these PRRs orchestrate antifungal immunity and briefly discuss progress in the use of these receptors as targets for antifungal and other vaccines.
Collapse
Affiliation(s)
- Sarah E Hardison
- Aberdeen Fungal Group, Section of Immunology and Infection, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|