51
|
Lebouvier M, Miramón-Puértolas P, Steinmetz PRH. Evolutionarily conserved aspects of animal nutrient uptake and transport in sea anemone vitellogenesis. Curr Biol 2022; 32:4620-4630.e5. [PMID: 36084649 DOI: 10.1016/j.cub.2022.08.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
The emergence of systemic nutrient transport was a key challenge during animal evolution, yet it is poorly understood. Circulatory systems distribute nutrients in many bilaterians (e.g., vertebrates and arthropods) but are absent in non-bilaterians (e.g., cnidarians and sponges), where nutrient absorption and transport remain little explored at molecular and cellular levels. Vitellogenesis, the accumulation of egg yolk, necessitates high nutrient influx into oocytes and is present throughout animal phyla and therefore represents a well-suited paradigm to study nutrient transport evolution. With that aim, we investigated dietary nutrient transport to the oocytes in the cnidarian Nematostella vectensis (Anthozoa). Using a combination of fluorescent bead labeling and marker gene expression, we found that phagocytosis, micropinocytosis, and intracellular digestion of food components occur within the gonad epithelium. Pulse-chase experiments further show that labelled fatty acids rapidly translocate from the gonad epithelium through the extracellular matrix (ECM) into oocytes. Expression of conserved lipid transport proteins vitellogenin (vtg) and apolipoprotein-B (apoB) and colocalization of labeled fatty acids with a fluorescently tagged ApoB protein further support the lipid-shuttling role of the gonad epithelium. Complementary oocyte expression of very low-density lipoprotein receptor (vldlr) orthologs, which mediate endocytosis of bilaterian ApoB- and Vtg-lipoproteins, supports that this evolutionarily conserved ligand/receptor pair underlies lipid transport during sea anemone vitellogenesis. In addition, we identified lipid- and ApoB-rich cells with potential lipid transport roles in the ECM. Altogether, our work supports a long-standing hypothesis that an ECM-based lipid transport system predated the cnidarian-bilaterian split and provided a basis for the evolution of bilaterian circulatory systems.
Collapse
Affiliation(s)
- Marion Lebouvier
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008 Bergen, Norway
| | - Paula Miramón-Puértolas
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008 Bergen, Norway
| | - Patrick R H Steinmetz
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008 Bergen, Norway.
| |
Collapse
|
52
|
Microglial Dynamics Modulate Vestibular Compensation in a Rodent Model of Vestibulopathy and Condition the Expression of Plasticity Mechanisms in the Deafferented Vestibular Nuclei. Cells 2022; 11:cells11172693. [PMID: 36078101 PMCID: PMC9454928 DOI: 10.3390/cells11172693] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Unilateral vestibular loss (UVL) induces a vestibular syndrome composed of posturo-locomotor, oculomotor, vegetative, and perceptivo-cognitive symptoms. With time, these functional deficits progressively disappear due to a phenomenon called vestibular compensation, known to be supported by the expression in the deafferented vestibular nuclei (VNs) of various adaptative plasticity mechanisms. UVL is known to induce a neuroinflammatory response within the VNs, thought to be caused by the structural alteration of primary vestibular afferents. The acute inflammatory response, expressed in the deafferented VNs was recently proven to be crucial for the expression of the endogenous plasticity supporting functional recovery. Neuroinflammation is supported by reactive microglial cells, known to have various phenotypes with adverse effects on brain tissue. Here, we used markers of pro-inflammatory and anti-inflammatory phenotypes of reactive microglia to study microglial dynamics following a unilateral vestibular neurectomy (UVN) in the adult rat. In addition, to highlight the role of acute inflammation in vestibular compensation and its underlying mechanisms, we enhanced the inflammatory state of the deafferented VNs using systemic injections of lipopolysaccharide (LPS) during the acute phase after a UVN. We observed that the UVN induced the expression of both M1 proinflammatory and M2 anti-inflammatory microglial phenotypes in the deafferented VNs. The acute LPS treatment exacerbated the inflammatory reaction and increased the M1 phenotype while decreasing M2 expression. These effects were associated with impaired postlesional plasticity in the deafferented VNs and exacerbated functional deficits. These results highlight the importance of a homeostatic inflammatory level in the expression of the adaptative plasticity mechanisms underlying vestibular compensation. Understanding the rules that govern neuroinflammation would provide therapeutic leads in neuropathologies associated with these processes.
Collapse
|
53
|
Pneumolysin boosts the neuroinflammatory response to Streptococcus pneumoniae through enhanced endocytosis. Nat Commun 2022; 13:5032. [PMID: 36028511 PMCID: PMC9418233 DOI: 10.1038/s41467-022-32624-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
In pneumococcal meningitis, bacterial growth in the cerebrospinal fluid results in lysis, the release of toxic factors, and subsequent neuroinflammation. Exposure of primary murine glia to Streptococcus pneumoniae lysates leads to strong proinflammatory cytokine and chemokine production, blocked by inhibition of the intracellular innate receptor Nod1. Lysates enhance dynamin-dependent endocytosis, and dynamin inhibition reduces neuroinflammation, blocking ligand internalization. Here we identify the cholesterol-dependent cytolysin pneumolysin as a pro-endocytotic factor in lysates, its elimination reduces their proinflammatory effect. Only pore-competent pneumolysin enhances endocytosis in a dynamin-, phosphatidylinositol-3-kinase- and potassium-dependent manner. Endocytic enhancement is limited to toxin-exposed parts of the membrane, the effect is rapid and pneumolysin permanently alters membrane dynamics. In a murine model of pneumococcal meningitis, mice treated with chlorpromazine, a neuroleptic with a complementary endocytosis inhibitory effect show reduced neuroinflammation. Thus, the dynamin-dependent endocytosis emerges as a factor in pneumococcal neuroinflammation, and its enhancement by a cytolysin represents a proinflammatory control mechanism.
Collapse
|
54
|
Synthesis and Immunological Evaluation of Mannosylated Desmuramyl Dipeptides Modified by Lipophilic Triazole Substituents. Int J Mol Sci 2022; 23:ijms23158628. [PMID: 35955759 PMCID: PMC9368957 DOI: 10.3390/ijms23158628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
Muramyl dipeptide (N-acetylmuramyl-L-alanyl-D-isoglutamine, MDP) is the smallest peptidoglycan fragment able to trigger an immune response by activating the NOD2 receptor. Structural modification of MDP can lead to analogues with improved immunostimulating properties. The aim of this work was to prepare mannosylated desmuramyl peptides (ManDMP) containing lipophilic triazole substituents to study their immunomodulating activities in vivo. The adjuvant activity of the prepared compounds was evaluated in the mouse model using ovalbumin as an antigen and compared to the MDP and referent adjuvant ManDMPTAd. The obtained results confirm that the α-position of D-isoGln is the best position for the attachment of lipophilic substituents, especially adamantylethyl triazole. Compound 6c exhibited the strongest adjuvant activity, comparable to the MDP and better than referent ManDMPTAd.
Collapse
|
55
|
Liu R, Qi Y, Zhai Y, Li H, An L, Yang G, Shan S. Identification and functional analysis of Mannose receptor in Asian swamp eel (Monopterus albus) in response to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2022; 127:463-473. [PMID: 35781053 DOI: 10.1016/j.fsi.2022.06.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/18/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Mannose receptor (MR), as a member of the C-type lectin (CLEC) family, plays an important role in the internalize pathogen-associated ligands and activate immune response. In the present study, MR was identified and characterized from Asian swamp eel (Monopterus albus) (namely MaMR). The open reading frame of MaMR was 4311 bp in length encoding 1437 amino acids of a ∼162.308 kDa protein, including a cysteine-rich (CR) domain, a fibronectin type II (FNII) domain, eight C-type lectin-like domains (CTLDs), a transmembrane domain and a short cytoplasmic domain. Phylogenetic analysis indicated that MaMR shared the highest similarity with that of Paralichthys olivaceus. The expression of MaMR was found in all the examined tissues, with the highest expression in the spleen and kidney. After injection with Edwardsiella tarda, the transcript level of MaMR was initially reduced and then significantly elevated in the liver, spleen, foregut and hindgut. In the isolated peripheral blood leukocytes, the expression of MaMR was significantly induced post stimulated with LPS and LTA. Then the MaMR-CTLD4-8 recombinant protein was purified. Bacterial agglutination and binding assay showed that rMaMR-CTLD4-8 could bind with both Gram-positive and Gram-negative bacteria and agglutinate bacteria in the presence of calcium in vitro. Further analysis revealed that MaMR and TLR2 coordinately induced the expression of TRAF6 and promoted the phosphorylation level of p65, leading to the expression of proinflammatory cytokines il-1β and tnf-α in EPC cells. Taken together, these results reveal that MaMR plays an important role in the immune response of fish to pathogen infections.
Collapse
Affiliation(s)
- Rongrong Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China
| | - Yue Qi
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China
| | - Yaqing Zhai
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China
| | - Liguo An
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China.
| | - Shijuan Shan
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No.88 East Wenhua Road, Jinan, 250014, China.
| |
Collapse
|
56
|
Fresquet M, Lockhart-Cairns MP, Rhoden SJ, Jowitt TA, Briggs DC, Baldock C, Brenchley PE, Lennon R. Structure of PLA2R reveals presentation of the dominant membranous nephropathy epitope and an immunogenic patch. Proc Natl Acad Sci U S A 2022; 119:e2202209119. [PMID: 35858348 PMCID: PMC9303975 DOI: 10.1073/pnas.2202209119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/31/2022] [Indexed: 01/04/2023] Open
Abstract
Membranous nephropathy is an autoimmune kidney disease caused by autoantibodies targeting antigens present on glomerular podocytes, instigating a cascade leading to glomerular injury. The most prevalent circulating autoantibodies in membranous nephropathy are against phospholipase A2 receptor (PLA2R), a cell surface receptor. The dominant epitope in PLA2R is located within the cysteine-rich domain, yet high-resolution structure-based mapping is lacking. In this study, we define the key nonredundant amino acids in the dominant epitope of PLA2R involved in autoantibody binding. We further describe two essential regions within the dominant epitope and spacer requirements for a synthetic peptide of the epitope for drug discovery. In addition, using cryo-electron microscopy, we have determined the high-resolution structure of PLA2R to 3.4 Å resolution, which shows that the dominant epitope and key residues within the cysteine-rich domain are accessible at the cell surface. In addition, the structure of PLA2R not only suggests a different orientation of domains but also implicates a unique immunogenic signature in PLA2R responsible for inducing autoantibody formation and recognition.
Collapse
Affiliation(s)
- Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Michael P. Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Samuel J. Rhoden
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Thomas A. Jowitt
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David C. Briggs
- Signalling and Structural Biology Lab, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
| | - Paul E. Brenchley
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, M13 9PT, United Kingdom
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, United Kingdom
- Department of Paediatric Nephrology, Royal Manchester Children’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| |
Collapse
|
57
|
Rastall RA, Diez-Municio M, Forssten SD, Hamaker B, Meynier A, Moreno FJ, Respondek F, Stah B, Venema K, Wiese M. Structure and function of non-digestible carbohydrates in the gut microbiome. Benef Microbes 2022; 13:95-168. [PMID: 35729770 DOI: 10.3920/bm2021.0090] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Together with proteins and fats, carbohydrates are one of the macronutrients in the human diet. Digestible carbohydrates, such as starch, starch-based products, sucrose, lactose, glucose and some sugar alcohols and unusual (and fairly rare) α-linked glucans, directly provide us with energy while other carbohydrates including high molecular weight polysaccharides, mainly from plant cell walls, provide us with dietary fibre. Carbohydrates which are efficiently digested in the small intestine are not available in appreciable quantities to act as substrates for gut bacteria. Some oligo- and polysaccharides, many of which are also dietary fibres, are resistant to digestion in the small intestines and enter the colon where they provide substrates for the complex bacterial ecosystem that resides there. This review will focus on these non-digestible carbohydrates (NDC) and examine their impact on the gut microbiota and their physiological impact. Of particular focus will be the potential of non-digestible carbohydrates to act as prebiotics, but the review will also evaluate direct effects of NDC on human cells and systems.
Collapse
Affiliation(s)
- R A Rastall
- Department of Food and Nutritional Sciences, The University of Reading, P.O. Box 226, Whiteknights, Reading, RG6 6AP, United Kingdom
| | - M Diez-Municio
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - S D Forssten
- IFF Health & Biosciences, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - B Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, 745 Agriculture Mall Drive, West Lafayette, IN 47907-2009, USA
| | - A Meynier
- Nutrition Research, Mondelez France R&D SAS, 6 rue René Razel, 91400 Saclay, France
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - F Respondek
- Tereos, Zoning Industriel Portuaire, 67390 Marckolsheim, France
| | - B Stah
- Human Milk Research & Analytical Science, Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands.,Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands
| | - K Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University - campus Venlo, St. Jansweg 20, 5928 RC Venlo, the Netherlands
| | - M Wiese
- Department of Microbiology and Systems Biology, TNO, Utrechtseweg 48, 3704 HE, Zeist, the Netherlands
| |
Collapse
|
58
|
Yin Z, Nie H, Jiang K, Yan X. Molecular Mechanisms Underlying Vibrio Tolerance in Ruditapes philippinarum Revealed by Comparative Transcriptome Profiling. Front Immunol 2022; 13:879337. [PMID: 35615362 PMCID: PMC9125321 DOI: 10.3389/fimmu.2022.879337] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022] Open
Abstract
The clam Ruditapes philippinarum is an important species in the marine aquaculture industry in China. However, in recent years, the aquaculture of R. philippinarum has been negatively impacted by various bacterial pathogens. In this study, the transcriptome libraries of R. philippinarum showing different levels of resistance to challenge with Vibrio anguillarum were constructed and RNA-seq was performed using the Illumina sequencing platform. Host immune factors were identified that responded to V. anguillarum infection, including C-type lectin domain, glutathione S-transferase 9, lysozyme, methyltransferase FkbM domain, heat shock 70 kDa protein, Ras-like GTP-binding protein RHO, C1q, F-box and BTB/POZ domain protein zf-C2H2. Ten genes were selected and verified by RT-qPCR, and nine of the gene expression results were consistent with those of RNA-seq. The lectin gene in the phagosome pathway was expressed at a significantly higher level after V. anguillarum infection, which might indicate the role of lectin in the immune response to V. anguillarum. Comparing the results from R. philippinarum resistant and nonresistant to V. anguillarum increases our understanding of the resistant genes and key pathways related to Vibrio challenge in this species. The results obtained here provide a reference for future immunological research focusing on the response of R. philippinarum to V. anguillarum infection.
Collapse
Affiliation(s)
- Zhihui Yin
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Hongtao Nie
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Kunyin Jiang
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xiwu Yan
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| |
Collapse
|
59
|
Ruan S, Huang Y, He M, Gao H. Advanced Biomaterials for Cell-Specific Modulation and Restore of Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200027. [PMID: 35343112 PMCID: PMC9165523 DOI: 10.1002/advs.202200027] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/18/2022] [Indexed: 05/09/2023]
Abstract
The past decade has witnessed the explosive development of cancer immunotherapies. Nevertheless, low immunogenicity, limited specificity, poor delivery efficiency, and off-target side effects remain to be the major limitations for broad implementation of cancer immunotherapies to patient bedside. Encouragingly, advanced biomaterials offering cell-specific modulation of immunological cues bring new solutions for improving the therapeutic efficacy while relieving side effect risks. In this review, focus is given on how functional biomaterials can enable cell-specific modulation of cancer immunotherapy within the cancer-immune cycle, with particular emphasis on antigen-presenting cells (APCs), T cells, and tumor microenvironment (TME)-resident cells. By reviewing the current progress in biomaterial-based cancer immunotherapy, here the aim is to provide a better understanding of biomaterials' role in targeting modulation of antitumor immunity step-by-step and guidelines for rationally developing targeting biomaterials for more personalized cancer immunotherapy. Moreover, the current challenge and future perspective regarding the potential application and clinical translation will also be discussed.
Collapse
Affiliation(s)
- Shaobo Ruan
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Mei He
- College of PharmacyUniversity of FloridaGainesvilleFL32610USA
| | - Huile Gao
- West China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
60
|
Inhalable Mannosylated Rifampicin–Curcumin Co-Loaded Nanomicelles with Enhanced In Vitro Antimicrobial Efficacy for an Optimized Pulmonary Tuberculosis Therapy. Pharmaceutics 2022; 14:pharmaceutics14050959. [PMID: 35631546 PMCID: PMC9145552 DOI: 10.3390/pharmaceutics14050959] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Among respiratory infections, tuberculosis was the second deadliest infectious disease in 2020 behind COVID-19. Inhalable nanocarriers offer the possibility of actively targeting anti-tuberculosis drugs to the lungs, especially to alveolar macrophages (cellular reservoirs of the Mycobacterium tuberculosis). Our strategy was based on the development of a mannose-decorated micellar nanoformulation based in Soluplus® to co-encapsulate rifampicin and curcumin. The former is one of the most effective anti-tuberculosis first-line drugs, while curcumin has demonstrated potential anti-mycobacterial properties. Mannose-coated rifampicin (10 mg/mL)–curcumin (5 mg/mL)-loaded polymeric micelles (10% w/v) demonstrated excellent colloidal properties with micellar size ~108 ± 1 nm after freeze-drying, and they remain stable under dilution in simulated interstitial lung fluid. Drug-loaded polymeric micelles were suitable for drug delivery to the deep lung with lung accumulation, according to the in vitro nebulization studies and the in vivo biodistribution assays of radiolabeled (99mTc) polymeric micelles, respectively. Hence, the nanoformulation did not exhibit hemolytic potential. Interestingly, the addition of mannose significantly improved (5.2-fold) the microbicidal efficacy against Mycobacterium tuberculosis H37Rv of the drug-co-loaded systems in comparison with their counterpart mannose-free polymeric micelles. Thus, this novel inhaled nanoformulation has demonstrated its potential for active drug delivery in pulmonary tuberculosis therapy.
Collapse
|
61
|
Paredes-Rojas A, Palma-Ramos A, Castrillón-Rivera LE, Mendoza-Pérez F, Navarro-González MDC, Arenas-Guzmán R, Castañeda-Sánchez JI, Luna-Herrera J. Keratinocyte Response to Infection with Sporothrix schenckii. J Fungi (Basel) 2022; 8:jof8050437. [PMID: 35628694 PMCID: PMC9143681 DOI: 10.3390/jof8050437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/27/2022] Open
Abstract
Sporotrichosis is a subacute, or chronic mycosis caused by traumatic inoculation of material contaminated with the fungus Sporothrix schenckii which is part of the Sporothrix spp. complex. The infection is limited to the skin, although its progression to more severe systemic or disseminated forms remains possible. Skin is the tissue that comes into contact with Sporothrix first, and the role of various cell lines has been described with regard to infection control. However, there is little information on the response of keratinocytes. In this study, we used the human keratinocyte cell line (HaCaT) and evaluated different aspects of infection from modifications in the cytoskeleton to the expression of molecules of the innate response during infection with conidia and yeast cells of Sporothrix schenckii. We found that during infection with both phases of the fungus, alterations of the actin cytoskeleton, formation of membrane protuberances, and loss of stress fibers were induced. We also observed an overexpression of the surface receptors MR, TLR6, CR3 and TLR2. Cytokine analysis showed that both phases of the fungus induced the production of elevated levels of the chemokines MCP-1 and IL-8, and proinflammatory cytokines IFN-α, IFN-γ and IL-6. In contrast, TNF-α production was significant only with conidial infection. In late post-infection, cytokine production was observed with immunoregulatory activity, IL-10, and growth factors, G-CSF and GM-CSF. In conclusion, infection of keratinocytes with conidia and yeast cells of Sporothrix schenckii induces an inflammatory response and rearrangements of the cytoskeleton.
Collapse
Affiliation(s)
- Araceli Paredes-Rojas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Alejandro Palma-Ramos
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Laura Estela Castrillón-Rivera
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
| | - María del Carmen Navarro-González
- Laboratorio de Investigación en Enfermedades Reumáticas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Roberto Arenas-Guzmán
- Sección de Micología, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico;
| | - Jorge Ismael Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City 04960, Mexico; (A.P.-R.); (L.E.C.-R.); (F.M.-P.)
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| | - Julieta Luna-Herrera
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Correspondence: (J.I.C.-S.); (J.L.-H.); Tel.: +52-55-54-83-70-00 (ext. 2803) (J.I.C.-S.); +52-55-57-29-63-00 (ext. 62371) (J.L.-H.)
| |
Collapse
|
62
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
63
|
Eld HMS, Johnsen PR, Nielsen EM, Jørgensen FZ, Lindstrøm-Svendsen M, Baldry M, Ingmer H, Frøkiær H. Soluble C-Type Lectin-Receptor Ligands Stimulate ROS Production in Dendritic Cells and Potentiate Killing of MRSA as Well as the MRSA Induced IL-12 Production. Front Immunol 2022; 13:845881. [PMID: 35386713 PMCID: PMC8977849 DOI: 10.3389/fimmu.2022.845881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/24/2022] [Indexed: 12/04/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) has developed resistance to most β-lactam antibiotics leaving few treatment options against infections with MRSA. Through mannose receptors, mannan potentiates IL-12 production induced by Gram-positive bacteria, a cytokine crucial in the clearance of S. aureus infection. We investigated the IL-12 potentiating effect of mannan pre-treatment of bone marrow-derived dendritic cells prior to stimulation with clinical MRSA strains. Mannan almost doubled IL-12 as well as IFN-β production in response to USA300, also when USA300 was treated with the β-lactam cefoxitin. The MRSA-induced IL-12 production was dependent on bacterial uptake and reactive oxygen species (ROS). Mannan alone induced ROS production, and in combination with USA300, the ROS produced corresponded to the sum induced by mannan and USA300. Addition of a monoclonal antibody against the mannose receptor likewise enhanced USA300-induced IL-12 and induced ROS production. Mannan addition further increased the endocytosis as well as the rate of endosomal killing of bacteria. Pre-treatment with soluble β-glucans also induced ROS and potentiated the USA300-induced IL-12 indicating that other C-type receptors may play a similar role. In the presence of the pro-inflammatory mediators, GM-CSF or IFN-γ, the mannan-enhanced IL-12 production increased further. The USA300-induced and the mannan-facilitated enhanced IFN-β and IL-12 showed same dependency on MAPK c-Jun N-terminal kinase signaling, suggesting that mannan enhances the signals already induced by the bacteria, rather than changing them. We suggest that the C-type lectin-induced ROS production is a key factor in the IFN-β and IL-12 potentiation.
Collapse
Affiliation(s)
- Helene M S Eld
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter R Johnsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Emilie M Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederikke Z Jørgensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Mara Baldry
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Frøkiær
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
64
|
Liu Z, Li Y, Li C, Lei G, Zhou L, Chen X, Jia X, Lu Y. Intestinal Candida albicans Promotes Hepatocarcinogenesis by Up-Regulating NLRP6. Front Microbiol 2022; 13:812771. [PMID: 35369462 PMCID: PMC8964356 DOI: 10.3389/fmicb.2022.812771] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a primary liver cancer, is closely associated with the gut microbiota. However, the role of gut fungi in the development of HCC remains unclear. The aim of this study was to explore the influence of intestinal Candida albicans on HCC. Here, We found that patients with HCC showed significantly decreased diversity of the gut mycobiome and increased abundance of C. albicans, compared to the patients with liver cirrhosis. The gavage of C. albicans in the WT models increased the tumor size and weight and influenced the plasma metabolome, which was indicated by alterations in 117 metabolites, such as L-carnitine and L-acetylcarnitine, and several KEGG enriched pathways, such as phenylalanine metabolism and citrate cycle. Moreover, the expression of nucleotide oligomerization domain-like receptor family pyrin domain containing 6 (NLRP6) in the intestinal tissues and primary intestinal epithelial cells of the WT mice interacted with C. albicans increased. Notably, the colonization of C. albicans had no effect on tumor growth in Nlrp6–/– mice. In conclusion, the abnormal colonization of C. albicans reprogrammed HCC metabolism and contributed to the progression of HCC dependent on NLRP6, which provided new targets for the treatment of HCC.
Collapse
Affiliation(s)
- Zherui Liu
- Peking University 302 Clinical Medical School, Beijing, China.,Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yinyin Li
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chen Li
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guanglin Lei
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lin Zhou
- Senior Department of Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiangling Chen
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaodong Jia
- Senior Department of Oncology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yinying Lu
- Peking University 302 Clinical Medical School, Beijing, China.,Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
65
|
Wang J, Wu CS, Hu YZ, Yang L, Zhang XJ, Zhang YA. Plasmablasts induced by chitosan oligosaccharide secrete natural IgM to enhance the humoral immunity in grass carp. Carbohydr Polym 2022; 281:119073. [DOI: 10.1016/j.carbpol.2021.119073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
|
66
|
Yu W, Shen L, Qi Q, Hu T. Conjugation with loxoribine and mannan improves the immunogenicity of Mycobacterium tuberculosis CFP10-TB10.4 fusion protein. Eur J Pharm Biopharm 2022; 172:193-202. [DOI: 10.1016/j.ejpb.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/01/2022] [Accepted: 02/15/2022] [Indexed: 11/04/2022]
|
67
|
Yin X, Bai H, Mu L, Chen N, Qi W, Huang Y, Xu H, Jian J, Wang A, Ye J. Expression and functional characterization of the mannose receptor (MR) from Nile tilapia (Oreochromis niloticus) in response to bacterial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104257. [PMID: 34530040 DOI: 10.1016/j.dci.2021.104257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/11/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
Mannose receptor (MR) as a member of the pattern recognition receptors (PRRs) plays an important role in the immune response. In mammals, the role of MR in the regulation of phagocytosis is clarified; however, its contribution to opsonize phagocytosis remains unclear in bony fish. In this study, the expression pattern of Nile tilapia mannose receptor gene (OnMR) was investigated and its regulation of the phagocytosis of monocytes/macrophages to pathogenic bacteria was identified. The full-length of OnMR open reading frame is 4314 bp, encoding a peptide containing 1437 amino acid residues. The deduced amino acid sequence revealed that OnMR contained a cysteine-rich domain, a fibronectin type II domain, multiple C-type lectin-like domains, a transmembrane domain and a short cytoplasmic domain. Tissue distribution analysis showed the OnMR transcripts was widely distribute in the ten detected tissues, and highly expressed in head kidney, hind kidney, intestine and spleen. After S. agalactiae and A. hydrophila infection, the expression of OnMR in head kidney and spleen increased significantly. Moreover, the expression of OnMR in MO/Mø were also upregulated post the infection of bacteria and mannose solutions in vitro. This suggested that MR, as a mannose receptor on macrophage surface, could respond strongly to the stimulation of pathogenic bacteria. In addition, the (r)OnMR protein could effectively bind and agglutinate S. agalactiae and A. hydrophila, and regulate the phagocytic ability of monocytes/macrophages to pathogenic bacteria. These results suggest that OnMR is involved in response against bacterial infection in Nile tilapia, and this study will help us better understand the function of MR in teleost fish.
Collapse
Affiliation(s)
- Xiaoxue Yin
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| | - Hao Bai
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Liangliang Mu
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| | - Nuo Chen
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Weiwei Qi
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Yu Huang
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Heyi Xu
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Jichang Jian
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang 524088, PR China
| | - Anli Wang
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China
| | - Jianmin Ye
- School of Life Sciences, South China Normal University, Institute of Modern Aquaculture Science and Engineering, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangzhou 510631, PR China.
| |
Collapse
|
68
|
Vogel A, Brunner JS, Hajto A, Sharif O, Schabbauer G. Lipid scavenging macrophages and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159066. [PMID: 34626791 DOI: 10.1016/j.bbalip.2021.159066] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Macrophages are professional phagocytes, indispensable for maintenance of tissue homeostasis and integrity. Depending on their resident tissue, macrophages are exposed to highly diverse metabolic environments. Adapted to their niche, they can contribute to local metabolic turnover through metabolite uptake, conversion, storage and release. Disturbances in tissue homeostasis caused by infection, inflammation or damage dramatically alter the local milieu, impacting macrophage activation status and metabolism. In the case of persisting stimuli, defective macrophage responses ensue, which can promote tissue damage and disease. Especially relevant herein are disbalances in lipid rich environments, where macrophages are crucially involved in lipid uptake and turnover, preventing lipotoxicity. Lipid uptake is to a large extent facilitated by macrophage expressed scavenger receptors that are dynamically regulated and important in many metabolic diseases. Here, we review the receptors mediating lipid uptake and summarize recent findings on their role in health and disease. We further highlight the underlying pathways driving macrophage lipid acquisition and their impact on myeloid metabolic remodelling.
Collapse
Affiliation(s)
- Andrea Vogel
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Julia Stefanie Brunner
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Alexander Hajto
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Omar Sharif
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria.
| | - Gernot Schabbauer
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria.
| |
Collapse
|
69
|
Jiang F, Ding Y, Tian Y, Yang R, Quan M, Tong Z, Zhang X, Luo D, Chi Z, Liu C. Hydrolyzed low-molecular-weight polysaccharide from Enteromorpha prolifera exhibits high anti-inflammatory activity and promotes wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112637. [DOI: 10.1016/j.msec.2021.112637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/27/2022]
|
70
|
Single-molecule imaging of glycan-lectin interactions on cells with Glyco-PAINT. Nat Chem Biol 2021; 17:1281-1288. [PMID: 34764473 DOI: 10.1038/s41589-021-00896-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 09/10/2021] [Indexed: 11/08/2022]
Abstract
Most lectins bind carbohydrate ligands with relatively low affinity, making the identification of optimal ligands challenging. Here we introduce a point accumulation in nanoscale topography (PAINT) super-resolution microscopy method to capture weak glycan-lectin interactions at the single-molecule level in living cells (Glyco-PAINT). Glyco-PAINT exploits weak and reversible sugar binding to directly achieve single-molecule detection and quantification in cells and is used to establish the relative kon and koff rates of a synthesized library of carbohydrate-based probes, as well as the diffusion coefficient of the receptor-sugar complex. Uptake of ligands correlates with their binding affinity and residence time to establish structure-function relations for various synthetic glycans. We reveal how sugar multivalency and presentation geometry can be optimized for binding and internalization. Overall, Glyco-PAINT represents a powerful approach to study weak glycan-lectin interactions on the surface of living cells, one that can be potentially extended to a variety of lectin-sugar interactions.
Collapse
|
71
|
Lu Y, Shi Y, You J. Strategy and clinical application of up-regulating cross presentation by DCs in anti-tumor therapy. J Control Release 2021; 341:184-205. [PMID: 34774890 DOI: 10.1016/j.jconrel.2021.11.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/20/2022]
Abstract
The cross presentation of exogenous antigen (Ag) by dendritic cells (DCs) facilitates a diversified mode of T-cell activation, orchestrates specific humoral and cellular immunity, and contributes to an efficient anti-tumor immune response. DCs-mediated cross presentation is subject to both intrinsic and extrinsic factors, including the homing and phenotype of DCs, the spatiotemporal trafficking and degradation kinetics of Ag, and multiple microenvironmental clues, with many details largely unexplored. Here, we systemically review the current mechanistic understanding and regulation strategies of cross presentation by heterogeneous DC populations. We also provide insights into the future exploitation of DCs cross presentation for a better clinical efficacy in anti-tumor therapy.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
72
|
van der Zande HJP, Nitsche D, Schlautmann L, Guigas B, Burgdorf S. The Mannose Receptor: From Endocytic Receptor and Biomarker to Regulator of (Meta)Inflammation. Front Immunol 2021; 12:765034. [PMID: 34721436 PMCID: PMC8551360 DOI: 10.3389/fimmu.2021.765034] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/27/2021] [Indexed: 01/27/2023] Open
Abstract
The mannose receptor is a member of the C-type lectin (CLEC) family, which can bind and internalize a variety of endogenous and pathogen-associated ligands. Because of these properties, its role in endocytosis as well as antigen processing and presentation has been studied intensively. Recently, it became clear that the mannose receptor can directly influence the activation of various immune cells. Cell-bound mannose receptor expressed by antigen-presenting cells was indeed shown to drive activated T cells towards a tolerogenic phenotype. On the other hand, serum concentrations of a soluble form of the mannose receptor have been reported to be increased in patients suffering from a variety of inflammatory diseases and to correlate with severity of disease. Interestingly, we recently demonstrated that the soluble mannose receptor directly promotes macrophage proinflammatory activation and trigger metaflammation. In this review, we highlight the role of the mannose receptor and other CLECs in regulating the activation of immune cells and in shaping inflammatory responses.
Collapse
Affiliation(s)
| | - Dominik Nitsche
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Laura Schlautmann
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Sven Burgdorf
- Cellular Immunology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
73
|
Gray LT, Raczy MM, Briquez PS, Marchell TM, Alpar AT, Wallace RP, Volpatti LR, Sasso MS, Cao S, Nguyen M, Mansurov A, Budina E, Watkins EA, Solanki A, Mitrousis N, Reda JW, Yu SS, Tremain AC, Wang R, Nicolaescu V, Furlong K, Dvorkin S, Manicassamy B, Randall G, Wilson DS, Kwissa M, Swartz MA, Hubbell JA. Generation of potent cellular and humoral immunity against SARS-CoV-2 antigens via conjugation to a polymeric glyco-adjuvant. Biomaterials 2021; 278:121159. [PMID: 34634664 PMCID: PMC8482845 DOI: 10.1016/j.biomaterials.2021.121159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022]
Abstract
The SARS-CoV-2 virus has caused an unprecedented global crisis, and curtailing its spread requires an effective vaccine which elicits a diverse and robust immune response. We have previously shown that vaccines made of a polymeric glyco-adjuvant conjugated to an antigen were effective in triggering such a response in other disease models and hypothesized that the technology could be adapted to create an effective vaccine against SARS-CoV-2. The core of the vaccine platform is the copolymer p(Man-TLR7), composed of monomers with pendant mannose or a toll-like receptor 7 (TLR7) agonist. Thus, p(Man-TLR7) is designed to target relevant antigen-presenting cells (APCs) via mannose-binding receptors and then activate TLR7 upon endocytosis. The p(Man-TLR7) construct is amenable to conjugation to protein antigens such as the Spike protein of SARS-CoV-2, yielding Spike-p(Man-TLR7). Here, we demonstrate Spike-p(Man-TLR7) vaccination elicits robust antigen-specific cellular and humoral responses in mice. In adult and elderly wild-type mice, vaccination with Spike-p(Man-TLR7) generates high and long-lasting titers of anti-Spike IgGs, with neutralizing titers exceeding levels in convalescent human serum. Interestingly, adsorbing Spike-p(Man-TLR7) to the depot-forming adjuvant alum amplified the broadly neutralizing humoral responses to levels matching those in mice vaccinated with formulations based off of clinically-approved adjuvants. Additionally, we observed an increase in germinal center B cells, antigen-specific antibody secreting cells, activated T follicular helper cells, and polyfunctional Th1-cytokine producing CD4+ and CD8+ T cells. We conclude that Spike-p(Man-TLR7) is an attractive, next-generation subunit vaccine candidate, capable of inducing durable and robust antibody and T cell responses.
Collapse
Affiliation(s)
- Laura T Gray
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Michal M Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Priscilla S Briquez
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Tiffany M Marchell
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, United States
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Rachel P Wallace
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Lisa R Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Maria Stella Sasso
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Elyse A Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, 60637, United States
| | - Nikolaos Mitrousis
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Joseph W Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Shann S Yu
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Andrew C Tremain
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, United States
| | - Ruyi Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Vlad Nicolaescu
- Department of Microbiology, Howard T. Ricketts Laboratory, University of Chicago, Chicago, IL, 60637, United States
| | - Kevin Furlong
- Department of Microbiology, Howard T. Ricketts Laboratory, University of Chicago, Chicago, IL, 60637, United States
| | - Steve Dvorkin
- Department of Microbiology, Howard T. Ricketts Laboratory, University of Chicago, Chicago, IL, 60637, United States
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, 52242, United States
| | - Glenn Randall
- Department of Microbiology, Howard T. Ricketts Laboratory, University of Chicago, Chicago, IL, 60637, United States
| | - D Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, 21231, United States
| | - Marcin Kwissa
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States
| | - Melody A Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States; Committee on Immunology, University of Chicago, Chicago, IL, 60637, United States; Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, United States; Ben May Department of Cancer Research, University of Chicago, Chicago, IL, 60637, United States.
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, United States; Committee on Immunology, University of Chicago, Chicago, IL, 60637, United States; Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, United States.
| |
Collapse
|
74
|
He Y, Yu W, Xiao L, Shen L, Qi J, Hu T. Conjugation of Zika virus EDIII with CRM 197, 8-arm PEG and mannan for development of an effective Zika virus vaccine. Int J Biol Macromol 2021; 190:713-721. [PMID: 34474053 DOI: 10.1016/j.ijbiomac.2021.08.177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/15/2023]
Abstract
Zika virus (ZIKV) induces neurological and autoimmune complications such as microcephaly and Guillain-Barre syndrome. Effective vaccines are necessary to prevent the ZIKV infection. E protein of ZIKV is responsible for virus attachment, entry, and fusion. The domain III of E protein (EDIII) contains the neutralizing epitopes and is ideal to act as an antigen for ZIKV vaccine. However, EDIII is poorly immunogenic. CRM197 is a carrier protein and can activate T helper cells for EDIII. Mannan is a ligand of TLR-4 or TLR-2. Eight-arm PEG can link multiple EDIII molecules in one entity. In the present study, EDIII was covalently conjugated with CRM197, 8-arm PEG and mannan to improve the immunogenicity of EDIII. The conjugate (CRM-EDIII-PM) elicited high EDIII-specific antibody titers in the BALB/c mice. Th1-type cytokines (IFN-γ and IL-2) and Th2-type cytokines (IL-5 and IL-10) were secreted at a marked level. Thus, CRM-EDIII-PM could stimulate potent humoral and cellular immune response to EDIII. The serum exposure of CRM-EDIII-PM to the immune system was prolonged. Moreover, CRM-EDIII-PM did not lead to apparent toxicity to the organs. Therefore, CRM-EDIII-PM was expected as a promising vaccine candidate for its ability to induce strong immune responses.
Collapse
Affiliation(s)
- Yunxia He
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Weili Yu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Lucheng Xiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Lijuan Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jinming Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Tao Hu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
75
|
Erban T, Klimov PB, Harant K, Talacko P, Nesvorna M, Hubert J. Label-free proteomic analysis reveals differentially expressed Wolbachia proteins in Tyrophagus putrescentiae: Mite allergens and markers reflecting population-related proteome differences. J Proteomics 2021; 249:104356. [PMID: 34438106 DOI: 10.1016/j.jprot.2021.104356] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022]
Abstract
Tyrophagus putrescentiae is an astigmatid mite of great economic, medical and veterinary importance. The microbiome, especially intracellular bacteria, may affect allergy/allergen expression. We targeted Wolbachia proteins, allergen comparisons and markers in Wolbachia-mite interactions in three mite populations. A decoy database was constructed by proteogenomics using the T. putrescentiae draft genome, Wolbachia transcriptome assembly and current T. putrescentiae-related sequences in GenBank. Among thousands of mite-derived proteins, 18 Wolbachia proteins were reliably identified. We suggest that peroxiredoxin, bacterioferritin, ankyrin repeat domain-containing protein and DegQ family serine endoprotease indicate a higher-level bacterium-bacterium-host interaction. We produced evidence that the host-Wolbachia interaction is modulated through pattern recognition receptors (PRRs), mannose-binding lectins/mannose receptors, the cholinergic anti-inflammatory pathway with TNF-α, and others. We observed Tyr p 3 suppression in mites with Wolbachia, linking trypsin to PRR modulation. Nine out of the 12 current WHO/IUIS official allergens were reliably identified, but the remaining three allergens, Tyr p 1, 8 and 35, were detected as only trace hits. This study provides numerous markers for further Wolbachia-host interaction research. For accuracy, mite allergens should be considered according to abundance in species, but mite populations/strains, as well as their microbiome structure, may be key factors. SIGNIFICANCE: The astigmatid mites occurring in homes are significant producers of allergens that are highly dangerous to humans and domesticated animals. Mites are tightly associated with microorganisms that affect their biology and consequently allergy signatures. Mite populations were found to be infected with certain intracellular bacteria, but some populations lacked an intracellular bacterium. Our previous research showed that some populations of Tyrophagus putrescentiae are infected with Wolbachia, but some populations host additional bacteria of interest. Thus, there are not only interactions between the mites and Wolbachia but also likely an additional level of interaction that can be found in the interaction between different bacteria in the mites. These "higher-level" signatures and consequences that bacteria affect, including allergen production, are not understood in mites. In this study, we identified Wolbachia-specific proteins in mites for the first time. This study provides Wolbachia- and mite-derived markers that can be clues for describing "higher-level" mite-bacterium-bacterium interactions. Indeed, the microbiome contribution to allergies can potentially be derived directly from bacterial proteins, especially if they are abundant.
Collapse
Affiliation(s)
- Tomas Erban
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia.
| | - Pavel B Klimov
- School of Natural Sciences, Bangor University, Bangor LL57 2 UW, UK; Institute of Biology, University of Tyumen, Pirogova 3, 625043 Tyumen, Russia
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia; Institute for Environmental Studies, Faculty of Science, Charles University, Benatska 2, Prague 2 CZ-128 01, Czechia
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science, Charles University, BIOCEV, Prumyslova 595, Vestec CZ-25242, Czechia
| | - Marta Nesvorna
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| | - Jan Hubert
- Crop Research Institute, Drnovska 507/73, Prague 6-Ruzyne CZ-16106, Czechia
| |
Collapse
|
76
|
Bhandari S, Larsen AK, McCourt P, Smedsrød B, Sørensen KK. The Scavenger Function of Liver Sinusoidal Endothelial Cells in Health and Disease. Front Physiol 2021; 12:757469. [PMID: 34707514 PMCID: PMC8542980 DOI: 10.3389/fphys.2021.757469] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this review is to give an outline of the blood clearance function of the liver sinusoidal endothelial cells (LSECs) in health and disease. Lining the hundreds of millions of hepatic sinusoids in the human liver the LSECs are perfectly located to survey the constituents of the blood. These cells are equipped with high-affinity receptors and an intracellular vesicle transport apparatus, enabling a remarkably efficient machinery for removal of large molecules and nanoparticles from the blood, thus contributing importantly to maintain blood and tissue homeostasis. We describe here central aspects of LSEC signature receptors that enable the cells to recognize and internalize blood-borne waste macromolecules at great speed and high capacity. Notably, this blood clearance system is a silent process, in the sense that it usually neither requires or elicits cell activation or immune responses. Most of our knowledge about LSECs arises from studies in animals, of which mouse and rat make up the great majority, and some species differences relevant for extrapolating from animal models to human are discussed. In the last part of the review, we discuss comparative aspects of the LSEC scavenger functions and specialized scavenger endothelial cells (SECs) in other vascular beds and in different vertebrate classes. In conclusion, the activity of LSECs and other SECs prevent exposure of a great number of waste products to the immune system, and molecules with noxious biological activities are effectively “silenced” by the rapid clearance in LSECs. An undesired consequence of this avid scavenging system is unwanted uptake of nanomedicines and biologics in the cells. As the development of this new generation of therapeutics evolves, there will be a sharp increase in the need to understand the clearance function of LSECs in health and disease. There is still a significant knowledge gap in how the LSEC clearance function is affected in liver disease.
Collapse
Affiliation(s)
- Sabin Bhandari
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Anett Kristin Larsen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Peter McCourt
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Karen Kristine Sørensen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
77
|
Design, Synthesis, and Biological Evaluation of Desmuramyl Dipeptides Modified by Adamantyl-1,2,3-triazole. Molecules 2021; 26:molecules26216352. [PMID: 34770761 PMCID: PMC8587862 DOI: 10.3390/molecules26216352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/17/2022] Open
Abstract
Muramyl dipeptide (MDP) is the smallest peptidoglycan fragment able to trigger the immune response. Structural modification of MDP can lead to the preparation of analogs with improved immunostimulant properties, including desmuramyl peptides (DMPs). The aim of this work was to prepare the desmuramyl peptide (L-Ala-D-Glu)-containing adamantyl-triazole moiety and its mannosylated derivative in order to study their immunomodulatory activities in vivo. The adjuvant activity of the prepared compounds was evaluated in a murine model using ovalbumin as an antigen, and compared to the reference adjuvant ManAdDMP. The results showed that the introduction of the lipophilic adamantyl-triazole moiety at the C-terminus of L-Ala-D-Glu contributes to the immunostimulant activity of DMP, and that mannosylation of DMP modified with adamantyl-triazole causes the amplification of its immunostimulant activity.
Collapse
|
78
|
Yang X, Lu D, Wang R, Lian Z, Lin Z, Zhuo J, Chen H, Yang M, Tan W, Yang M, Wei X, Wei Q, Zheng S, Xu X. Single-cell profiling reveals distinct immune phenotypes that contribute to ischaemia-reperfusion injury after steatotic liver transplantation. Cell Prolif 2021; 54:e13116. [PMID: 34469018 PMCID: PMC8488562 DOI: 10.1111/cpr.13116] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/05/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The discrepancy between supply and demand of organ has led to an increased utilization of steatotic liver for liver transplantation (LT). Hepatic steatosis, however, is a major risk factor for graft failure due to increased susceptibility to ischaemia-reperfusion (I/R) injury during transplantation. MATERIALS AND METHODS To assess the plasticity and phenotype of immune cells within the microenvironment of steatotic liver graft at single-cell level, single-cell RNA-sequencing (scRNA-Seq) was carried out on 23 675 cells from transplanted rat livers. Bioinformatic analyses and multiplex immunohistochemistry were performed to assess the functional properties, transcriptional regulation, phenotypic switching and cell-cell interactions of different cell subtypes. RESULTS We have identified 11 different cell types in transplanted livers and found that the highly complex ecosystem was shaped by myeloid-derived cell subsets that transit between different states and interact mutually. Notably, a pro-inflammatory phenotype of Kupffer cells (KCs) with high expression of colony-stimulating factor 3 (CSF3) that was enriched in transplanted steatotic livers was potentially participated in fatty graft injury. We have also detected a subset of dendritic cells (DCs) with highly expressing XCR1 that was correlated with CD8+ T cells, mediating the severer steatotic liver damage by I/R injury. CONCLUSIONS The findings of our study provide new insight into the mechanisms by which steatosis exacerbates liver damage from I/R injury. Interventions based on these observations create opportunities in attenuating fatty liver graft injury and expanding the donor pool.
Collapse
|
79
|
Trivedi A, Tercovich KG, Casbon AJ, Raber J, Lowell C, Noble-Haeusslein LJ. Neutrophil-specific deletion of Syk results in recruitment-independent stabilization of the barrier and a long-term improvement in cognitive function after traumatic injury to the developing brain. Neurobiol Dis 2021; 157:105430. [PMID: 34153467 PMCID: PMC11302380 DOI: 10.1016/j.nbd.2021.105430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/14/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
While traumatic brain injury (TBI) is the leading cause of death and disability in children, we have yet to identify those pathogenic events that determine the extent of recovery. Neutrophils are best known as "first responders" to sites of infection and trauma where they become fully activated, killing pathogens via proteases that are released during degranulation. However, this activational state may generate substantial toxicity in the young brain after TBI that is partially due to developmentally regulated inadequate antioxidant reserves. Neutrophil degranulation is triggered via a downstream signaling pathway that is dependent on spleen tyrosine kinase (Syk). To test the hypothesis that the activational state of neutrophils is a determinant of early pathogenesis and long-term recovery, we compared young, brain-injured conditional knockouts of Syk (sykf/fMRP8-cre+) to congenic littermates (sykf/f). Based upon flow cytometry, there was an extended recruitment of distinct leukocyte subsets, including Ly6G+/Ly6C- and Ly6G+/Ly6Cint, over the first several weeks post-injury which was similar between genotypes. Subsequent assessment of the acutely injured brain revealed a reduction in blood-brain barrier disruption to both high and low molecular weight dextrans and reactive oxygen species in sykf/fMRP8-cre+ mice compared to congenic littermates, and this was associated with greater preservation of claudin 5 and neuronal integrity, as determined by Western blot analyses. At adulthood, motor learning was less affected in brain-injured sykf/fMRP8-cre+ mice as compared to sykf/f mice. Performance in the Morris Water Maze revealed a robust improvement in hippocampal-dependent acquisition and short and long-term spatial memory retention in sykf/fMRP8-cre+ mice. Subsequent analyses of swim path lengths during hidden platform training and probe trials showed greater thigmotaxis in brain-injured sykf/f mice than sham sykf/f mice and injured sykf/fMRP8-cre+ mice. Our results establish the first mechanistic link between the activation state of neutrophils and long-term functional recovery after traumatic injury to the developing brain. These results also highlight Syk kinase as a novel therapeutic target that could be further developed for the brain-injured child.
Collapse
Affiliation(s)
- Alpa Trivedi
- Departments of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA; Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Kayleen G Tercovich
- Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amy Jo Casbon
- Departments of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Clifford Lowell
- Departments of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Linda J Noble-Haeusslein
- Departments of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Departments of Neurology and Psychology, The Dell Medical School and the College of Liberal Arts, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
80
|
Lukácsi S, Farkas Z, Saskői É, Bajtay Z, Takács-Vellai K. Conserved and Distinct Elements of Phagocytosis in Human and C. elegans. Int J Mol Sci 2021; 22:ijms22168934. [PMID: 34445642 PMCID: PMC8396242 DOI: 10.3390/ijms22168934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Endocytosis provides the cellular nutrition and homeostasis of organisms, but pathogens often take advantage of this entry point to infect host cells. This is counteracted by phagocytosis that plays a key role in the protection against invading microbes both during the initial engulfment of pathogens and in the clearance of infected cells. Phagocytic cells balance two vital functions: preventing the accumulation of cell corpses to avoid pathological inflammation and autoimmunity, whilst maintaining host defence. In this review, we compare elements of phagocytosis in mammals and the nematode Caenorhabditis elegans. Initial recognition of infection requires different mechanisms. In mammals, pattern recognition receptors bind pathogens directly, whereas activation of the innate immune response in the nematode rather relies on the detection of cellular damage. In contrast, molecules involved in efferocytosis—the engulfment and elimination of dying cells and cell debris—are highly conserved between the two species. Therefore, C. elegans is a powerful model to research mechanisms of the phagocytic machinery. Finally, we show that both mammalian and worm studies help to understand how the two phagocytic functions are interconnected: emerging data suggest the activation of innate immunity as a consequence of defective apoptotic cell clearance.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Éva Saskői
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
- Department of Immunology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary
| | - Krisztina Takács-Vellai
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
- Correspondence:
| |
Collapse
|
81
|
Ouyang A, Wang H, Su J, Liu X. Mannose Receptor Mediates the Activation of Chitooligosaccharides on Blunt Snout Bream ( Megalobrama amblycephala) Macrophages. Front Immunol 2021; 12:686846. [PMID: 34408745 PMCID: PMC8365301 DOI: 10.3389/fimmu.2021.686846] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/16/2021] [Indexed: 01/23/2023] Open
Abstract
Chitooligosaccharide (COS) is an important immune enhancer and has been proven to have a variety of biological activities. Our previous research has established an M1 polarization mode by COS in blunt snout bream (Megalobrama amblycephala) macrophages, but the mechanism of COS activation of blunt snout bream macrophages remains unclear. In this study, we further explored the internalization mechanism and signal transduction pathway of chitooligosaccharide hexamer (COS6) in blunt snout bream macrophages. The results showed that mannose receptor C-type lectin-like domain 4-8 of M. amblycephala (MaMR CTLD4-8) could recognize and bind to COS6 and mediate COS6 into macrophages by both clathrin-dependent and caveolin-dependent pathways. In the inflammatory response of macrophages activated by COS6, the gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide synthase 2 (NOS2) was significantly inhibited after MaMR CTLD4-8-specific antibody blockade. However, even if it was blocked, the expression of these inflammation-related genes was still relatively upregulated, which suggested that there are other receptors involved in immune regulation. Further studies indicated that MaMR CTLD4-8 and Toll-like receptor 4 (TLR4) cooperated to regulate the pro-inflammatory response of macrophages caused by COS6. Taken together, these results revealed that mannose receptor (MR) CTLD4-8 is indispensable in the process of recognition, binding, internalization, and immunoregulation of COS in macrophages of blunt snout bream.
Collapse
Affiliation(s)
- Aotian Ouyang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Huabing Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| |
Collapse
|
82
|
Leuti A, Talamonti E, Gentile A, Tiberi M, Matteocci A, Fresegna D, Centonze D, Chiurchiù V. Macrophage Plasticity and Polarization Are Altered in the Experimental Model of Multiple Sclerosis. Biomolecules 2021; 11:biom11060837. [PMID: 34200023 PMCID: PMC8229971 DOI: 10.3390/biom11060837] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/09/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system. MS is characterized by infiltrations of leukocytes such as T and B lymphocytes and macrophages. Macrophages have been identified as major effectors of inflammation and demyelination in both MS and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the activation and heterogeneity of macrophages in MS has been poorly investigated. Thus, in this study, we evaluated M1 and M2 macrophages immunophenotype from EAE and control mice by analyzing over 30 surface and intracellular markers through polychromatic flow cytometry, qRT-PCR, and ELISA assay. We showed that M1 macrophages possessed a higher proinflammatory profile in EAE compared to control mice, since they expressed higher levels of activation/co-stimulatory markers (iNOS, CD40, and CD80) and cytokines/chemokines (IL-6, IL-12, CCL2, and CXCL10), whereas M2 lost their M2-like phenotype by showing a decreased expression of their signature markers CD206 and CCL22, as well as a concomitant upregulation of several M1 makers. Furthermore, immunization of M1 and M2 macrophages with MOG35-55 led to a significant hyperactivation of M1 and a concomitant shift of anti-inflammatory M2 to pro-inflammatory M1 macrophages. Overall, we provide evidence for a phenotypic alteration of M1/M2 balance during MS, which can be of crucial importance not only for a better understanding of the immunopathology of this neurodegenerative disease but also to potentially develop new macrophage-centered therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Leuti
- Department of Medicine, Campus Bio-Medico University of Rome, 00128 Rome, Italy;
- Laboratory of Neurochemistry of Lipids, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Emanuela Talamonti
- Department of Molecular Biosciences, The Wenner-Gren Institute, University of Stockholm, 114 Stockholm, Sweden;
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.G.); (D.F.)
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (M.T.); (A.M.)
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (M.T.); (A.M.)
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00163 Rome, Italy; (A.G.); (D.F.)
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy;
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy;
- Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (M.T.); (A.M.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
- Correspondence: or ; Tel.: +39-06-501703210
| |
Collapse
|
83
|
Luque-Campos N, Bustamante-Barrientos FA, Pradenas C, García C, Araya MJ, Bohaud C, Contreras-López R, Elizondo-Vega R, Djouad F, Luz-Crawford P, Vega-Letter AM. The Macrophage Response Is Driven by Mesenchymal Stem Cell-Mediated Metabolic Reprogramming. Front Immunol 2021; 12:624746. [PMID: 34149687 PMCID: PMC8213396 DOI: 10.3389/fimmu.2021.624746] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stromal cells widely studied for their regenerative and immunomodulatory properties. They are capable of modulating macrophage plasticity depending on various microenvironmental signals. Current studies have shown that metabolic changes can also affect macrophage fate and function. Indeed, changes in the environment prompt phenotype change. Therefore, in this review, we will discuss how MSCs orchestrate macrophage’s metabolic plasticity and the impact on their function. An improved understanding of the crosstalk between macrophages and MSCs will improve our knowledge of MSC’s therapeutic potential in the context of inflammatory diseases, cancer, and tissue repair processes in which macrophages are pivotal.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | | | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
84
|
Botwright NA, Mohamed AR, Slinger J, Lima PC, Wynne JW. Host-Parasite Interaction of Atlantic salmon ( Salmo salar) and the Ectoparasite Neoparamoeba perurans in Amoebic Gill Disease. Front Immunol 2021; 12:672700. [PMID: 34135900 PMCID: PMC8202022 DOI: 10.3389/fimmu.2021.672700] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Marine farmed Atlantic salmon (Salmo salar) are susceptible to recurrent amoebic gill disease (AGD) caused by the ectoparasite Neoparamoeba perurans over the growout production cycle. The parasite elicits a highly localized response within the gill epithelium resulting in multifocal mucoid patches at the site of parasite attachment. This host-parasite response drives a complex immune reaction, which remains poorly understood. To generate a model for host-parasite interaction during pathogenesis of AGD in Atlantic salmon the local (gill) and systemic transcriptomic response in the host, and the parasite during AGD pathogenesis was explored. A dual RNA-seq approach together with differential gene expression and system-wide statistical analyses of gene and transcription factor networks was employed. A multi-tissue transcriptomic data set was generated from the gill (including both lesioned and non-lesioned tissue), head kidney and spleen tissues naïve and AGD-affected Atlantic salmon sourced from an in vivo AGD challenge trial. Differential gene expression of the salmon host indicates local and systemic upregulation of defense and immune responses. Two transcription factors, znfOZF-like and znf70-like, and their associated gene networks significantly altered with disease state. The majority of genes in these networks are candidates for mediators of the immune response, cellular proliferation and invasion. These include Aurora kinase B-like, rho guanine nucleotide exchange factor 25-like and protein NDNF-like inhibited. Analysis of the N. perurans transcriptome during AGD pathology compared to in vitro cultured N. perurans trophozoites, as a proxy for wild type trophozoites, identified multiple gene candidates for virulence and indicates a potential master regulatory gene system analogous to the two-component PhoP/Q system. Candidate genes identified are associated with invasion of host tissue, evasion of host defense mechanisms and formation of the mucoid lesion. We generated a novel model for host-parasite interaction during AGD pathogenesis through integration of host and parasite functional profiles. Collectively, this dual transcriptomic study provides novel molecular insights into the pathology of AGD and provides alternative theories for future research in a step towards improved management of AGD.
Collapse
Affiliation(s)
- Natasha A Botwright
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Amin R Mohamed
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Joel Slinger
- Livestock and Aquaculture, CSIRO Agriculture and Food, Woorim, QLD, Australia
| | - Paula C Lima
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - James W Wynne
- Livestock and Aquaculture, CSIRO Agriculture and Food, Hobart, TAS, Australia
| |
Collapse
|
85
|
FATP4 inactivation in cultured macrophages attenuates M1- and ER stress-induced cytokine release via a metabolic shift towards triacylglycerides. Biochem J 2021; 478:1861-1877. [PMID: 33900381 DOI: 10.1042/bcj20210155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Abstract
Fatty acid transport protein 4 (FATP4) belongs to a family of acyl-CoA synthetases which activate long-chain fatty acids into acyl-CoAs subsequently used in specific metabolic pathways. Patients with FATP4 mutations and Fatp4-null mice show thick desquamating skin and other complications, however, FATP4 role on macrophage functions has not been studied. We here determined whether the levels of macrophage glycerophospholipids, sphingolipids including ceramides, triacylglycerides, and cytokine release could be altered by FATP4 inactivation. Two in vitro experimental systems were studied: FATP4 knockdown in THP-1-derived macrophages undergoing M1 (LPS + IFNγ) or M2 (IL-4) activation and bone marrow-derived macrophages (BMDMs) from macrophage-specific Fatp4-knockout (Fatp4M-/-) mice undergoing tunicamycin (TM)-induced endoplasmic reticulum stress. FATP4-deficient macrophages showed a metabolic shift towards triacylglycerides and were protected from M1- or TM-induced release of pro-inflammatory cytokines and cellular injury. Fatp4M-/- BMDMs showed specificity in attenuating TM-induced activation of inositol-requiring enzyme1α, but not other unfolded protein response pathways. Under basal conditions, FATP4/Fatp4 deficiency decreased the levels of ceramides and induced an up-regulation of mannose receptor CD206 expression. The deficiency led to an attenuation of IL-8 release in THP-1 cells as well as TNF-α and IL-12 release in BMDMs. Thus, FATP4 functions as an acyl-CoA synthetase in macrophages and its inactivation suppresses the release of pro-inflammatory cytokines by shifting fatty acids towards the synthesis of specific lipids.
Collapse
|
86
|
Gottfried-Blackmore A, Namkoong H, Adler E, Martin B, Gubatan J, Fernandez-Becker N, Clarke JO, Idoyaga J, Nguyen L, Habtezion A. Gastric Mucosal Immune Profiling and Dysregulation in Idiopathic Gastroparesis. Clin Transl Gastroenterol 2021; 12:e00349. [PMID: 33979305 PMCID: PMC8132986 DOI: 10.14309/ctg.0000000000000349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION It is unclear how immune perturbations may influence the pathogenesis of idiopathic gastroparesis, a prevalent functional disorder of the stomach which lacks animal models. Several studies have noted altered immune characteristics in the deep gastric muscle layer associated with gastroparesis, but data are lacking for the mucosal layer, which is endoscopically accessible. We hypothesized that immune dysregulation is present in the gastroduodenal mucosa in idiopathic gastroparesis and that specific immune profiles are associated with gastroparesis clinical parameters. METHODS In this cross-sectional prospective case-control study, routine endoscopic biopsies were used for comprehensive immune profiling by flow cytometry, multicytokine array, and gene expression in 3 segments of the stomach and the duodenal bulb. Associations of immune endpoints with clinical parameters of gastroparesis were also explored. RESULTS The gastric mucosa displayed large regional variation of distinct immune profiles. Furthermore, several-fold increases in innate and adaptive immune cells were found in gastroparesis. Various immune cell types showed positive correlations with duration of disease, proton pump inhibitor dosing, and delayed gastric emptying. DISCUSSION This initial observational study showed immune compartmentalization of the human stomach mucosa and significant immune dysregulation at the level of leukocyte infiltration in idiopathic gastroparesis patients that extends to the duodenum. Select immune cells, such as macrophages, may correlate with clinicopathological traits of gastroparesis. This work supports further mucosal studies to advance our understanding of gastroparesis pathophysiology.
Collapse
Affiliation(s)
| | - Hong Namkoong
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Emerald Adler
- Northwestern University Feinberg School of Medicine,
Division of Gastroenterology and Hepatology, Chicago, Illinois, USA
| | - Brock Martin
- Department of Pathology, Stanford University,
Stanford, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Nielsen Fernandez-Becker
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - John O. Clarke
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford
University School of Medicine, Stanford, USA
| | - Linda Nguyen
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| |
Collapse
|
87
|
Villar J, Salazar ML, Jiménez JM, Campo MD, Manubens A, Gleisner MA, Ávalos I, Salazar-Onfray F, Salazar F, Mitchell DA, Alshahrani MY, Martínez-Pomares L, Becker MI. C-type lectin receptors MR and DC-SIGN are involved in recognition of hemocyanins, shaping their immunostimulatory effects on human dendritic cells. Eur J Immunol 2021; 51:1715-1731. [PMID: 33891704 DOI: 10.1002/eji.202149225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/22/2022]
Abstract
Hemocyanins are used as immunomodulators in clinical applications because they induce a strong Th1-biased cell-mediated immunity, which has beneficial effects. They are multiligand glycosylated molecules with abundant and complex mannose-rich structures. It remains unclear whether these structures influence hemocyanin-induced immunostimulatory processes in human APCs. We have previously shown that hemocyanin glycans from Concholepas concholepas (CCH), Fissurella latimarginata (FLH), and Megathura crenulata (KLH), participate in their immune recognition and immunogenicity in mice, interacting with murine C-type lectin receptors (CLRs). Here, we studied the interactions of these hemocyanins with two major mannose-binding CLRs on monocyte-derived human DCs: MR (mannose receptor) and DC-SIGN (DC-specific ICAM-3-grabbing nonintegrin). Diverse analyses showed that hemocyanins are internalized by a mannose-sensitive mechanism. This process was calcium dependent. Moreover, hemocyanins colocalized with MR and DC-SIGN, and were partly internalized through clathrin-mediated endocytosis. The hemocyanin-mediated proinflammatory cytokine response was impaired when using deglycosylated FLH and KLH compared to CCH. We further showed that hemocyanins bind to human MR and DC-SIGN in a carbohydrate-dependent manner with affinity constants in the physiological concentration range. Overall, we showed that these three clinically valuable hemocyanins interact with human mannose-sensitive CLRs, initiating an immune response and promoting a Th1 cell-driving potential.
Collapse
Affiliation(s)
- Javiera Villar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Michelle L Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - José M Jiménez
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Miguel Del Campo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Biosonda Corporation, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Daniel A Mitchell
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Biosonda Corporation, Santiago, Chile
| |
Collapse
|
88
|
Jambari NN, Liddell S, Martinez-Pomares L, Alcocer MJC. Effect of O-linked glycosylation on the antigenicity, cellular uptake and trafficking in dendritic cells of recombinant Ber e 1. PLoS One 2021; 16:e0249876. [PMID: 33914740 PMCID: PMC8084162 DOI: 10.1371/journal.pone.0249876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 03/29/2021] [Indexed: 11/18/2022] Open
Abstract
Ber e 1, a major Brazil nut allergen, has been successfully produced in the yeast Pichia pastoris expression system as homogenous recombinant Ber e 1 (rBer e 1) with similar physicochemical properties and identical immunoreactivity to its native counterpart, nBer e 1. However, O-linked glycans was detected on the P.pastoris-derived rBer e 1, which is not naturally present in nBer e 1, and may contribute to the allergic sensitisation. In this study, we addressed the glycosylation differences between P. pastoris-derived recombinant Ber e 1 and its native counterparts. We also determined whether this fungal glycosylation could affect the antigenicity and immunogenicity of the rBer e 1 by using dendritic cells (DC) as an immune cell model due to their role in modulating the immune response. We identified that the glycosylation occurs at Ser96, Ser101 and Ser110 on the large chain and Ser19 on the small polypeptide chain of rBer e 1 only. The glycosylation on rBer e 1 was shown to elicit varying degree of antigenicity by binding to different combination of human leukocyte antigens (HLA) at different frequencies compared to nBer e 1 when tested using human DC-T cell assay. However, both forms of Ber e 1 are weak immunogens based from their low response indexes (RI). Glycans present on rBer e 1 were shown to increase the efficiency of the protein recognition and internalization by murine bone marrow-derived dendritic cells (bmDC) via C-type lectin receptors, particularly the mannose receptor (MR), compared to the non-glycosylated nBer e 1 and SFA8, a weak allergenic 2S albumin protein from sunflower seed. Binding of glycosylated rBer e 1 to MR alone was found to not induce the production of IL-10 that modulates bmDC to polarise Th2 cell response by suppressing IL-12 production and DC maturation. Our findings suggest that the O-linked glycosylation by P. pastoris has a small but measurable effect on the in vitro antigenicity of the rBer e 1 compared to its non-glycosylated counterpart, nBer e 1, and thus may influence its applications in diagnostics and immunotherapy.
Collapse
Affiliation(s)
- Nuzul N. Jambari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Food Safety and Food Integrity (FOSFI), Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Susan Liddell
- Division of Animal Science, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Luisa Martinez-Pomares
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Marcos J. C. Alcocer
- Division of Food Sciences, School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| |
Collapse
|
89
|
Toribio RE, Young N, Schlesinger LS, Cope FO, Ralph DA, Jarjour W, Rosol TJ. Cy3-tilmanocept labeling of macrophages in joints of mice with antibody-induced arthritis and synovium of human patients with rheumatoid arthritis. J Orthop Res 2021; 39:821-830. [PMID: 33107629 DOI: 10.1002/jor.24900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 02/04/2023]
Abstract
γ-Tilmanocept (99m Tc-tilmanocept) is a receptor-directed, radiolabeled tracer that is FDA-approved for guiding sentinel lymph node biopsy. Tilmanocept binds the C-type lectin mannose receptor (MR, CD206) on macrophages. In this study, nonradioactive, fluorescently-labeled Cy3-tilmanocept was used to detect CD206+ mononuclear cells in the cartilage of mice with antibody-induced arthritis and in the synovial fluid and tissue of human subjects with rheumatoid arthritis (RA) for comparison with osteoarthritis (OA), and healthy volunteer (HV) controls. Murine arthritis was induced by injection of monoclonal anti-cartilage antibody followed by injection of Escherichia coli lipopolysaccharide. Post-arthritis development (7-11 days), the mice were injected intravenously with Cy3-tilmanocept followed by in vivo and ex vivo epifluorescence imaging. Two-photon imaging, immunofluorescence, and immunohistochemistry were used to identify articular and synovial macrophages (CD206, F4/80, and Cy3-tilmanocept binding) in murine tissues. Cy3-tilmanocept epifluorescence was present in arthritic knees and elbows of murine tissues; no radiographic changes were noted in the skeletons. However, inflammatory arthritic changes were apparent by histopathology and immunohistochemistry (F4/80), immunofluorescence (CD206) and Cy3-tilmanocept binding. In human RA synovial fluid, Cy3-tilmanocept staining correlated with CD206+ /CD16+ cells; negligible labeling was observed in OA samples. Cy3-tilmanocept colocalized with CD206 and staining was significantly higher in RA synovial tissue compared to OA or HV. Our results demonstrate that imaging with Cy3-tilmanocept can detect in vivo inflammatory, CD206+ macrophages in an early arthritis animal model and in human RA patients. These data establish a novel tool for preclinical research of early arthritis and have implications for early RA detection and monitoring of therapeutic efficacy in humans.
Collapse
Affiliation(s)
- Ramiro E Toribio
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Nicholas Young
- Division of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Larry S Schlesinger
- Department of Microbial Infection & Immunity, The Ohio State University, Columbus, Ohio, USA.,Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Fred O Cope
- Navidea Biopharmaceuticals, Inc., Dublin, Ohio, USA.,Physis International LLC, Westerville, Ohio, USA
| | | | - Wael Jarjour
- Division of Rheumatology and Immunology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Thomas J Rosol
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
90
|
Li Z, Wang J, Zhao Y, Ma D, Zhao M, Li N, Men Y, Zhang Y, Chu H, Lei C, Shen W, El-Mahdy Othman O, Min L. scRNA-seq of ovarian follicle granulosa cells from different fertility goats reveals distinct expression patterns. Reprod Domest Anim 2021; 56:801-811. [PMID: 33624340 DOI: 10.1111/rda.13920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 01/02/2023]
Abstract
The new technology of high-throughput single-cell RNA sequencing (10 × scRNA-seq) was developed recently with many advantages. However, it was not commonly used in farm animal research. There are few reports for the gene expression of goat ovarian follicle granulosa cells (GCs) during different developmental stages. In the current investigation, the gene expression of follicle GCs at different stages from two populations of Ji'ning grey goats: high litter size (HL; ≥3/L; 2 L) and low litter size (LL; ≤2 /L; 2 L) were analysed by scRNA-seq. Many GC marker genes were identified, and the pseudo-time showed that GCs developed during the time course which reflected the follicular development and differentiation trajectory. Moreover, the gene expression difference between the two populations HL versus LL was very clear at different developmental stages. Many marker genes differentially expressed at different developmental stages. ASIP and ASPN were found to be highly expressed in the early stage of GCs, INHA, INHBA, MFGE8 and HSD17B1 were identified to be highly expressed in the growing stage of GCs, while IGFBP2, IGFBP5 and CYP11A1 were found to be highly expressed in late stage. These marker genes could be used as reference genes of goat follicle GC development. This investigation for the first time discovered the gene expression patterns in goat follicle GCs in high- or low-fertility populations (based on litter size) by scRNA-seq which may be useful for uncovering the oocyte development potential.
Collapse
Affiliation(s)
- Zengkuan Li
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Junjie Wang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Yong Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Dongxue Ma
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Minghui Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Na Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Yuhao Men
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Yuan Zhang
- Jining Animal Husbandry Development Center, Jining, China
| | - Huimin Chu
- Jining Agricultural Science Institute, Jining, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | | | - Lingjiang Min
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
91
|
Liu E, Ji Y, Zhang F, Liu B, Meng X. Review on Auricularia auricula-judae as a Functional Food: Growth, Chemical Composition, and Biological Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1739-1750. [PMID: 33543932 DOI: 10.1021/acs.jafc.0c05934] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Although the application of Auricularia auricula-judae (AAJ) for health purposes has a long tradition in Asia, there is a lack of research on the functional nutrition of AAJ; the current research focused on polysaccharides has been too unitary compared to other mushrooms in recent years. Identification, extraction, and large-scale production of biologically active substances have emerged as critical determinants that determine AAJ becoming a functional food. AAJ is being treated in a restrained manner, despite having significant potential as a drug or a source of pure bioactive substances. Functional ingredients of mushrooms and AAJ have emerged as a new impetus for researchers interested in developing functional foods. This review presents an overview of current studies relevant to nutrition and the application of AAJ. The physiological conditions of AAJ and the corresponding functional ingredients beneficial to human health are reviewed to better understand the function and mechanisms of different nutrient contents. Relevant methods for evaluating the efficiency of extraction are also summarized. Finally, current limitations and the future scope for functional ingredients of AAJ are identified and discussed.
Collapse
Affiliation(s)
- Enchao Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Yuan Ji
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Fang Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Bingjie Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| | - Xianghong Meng
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, People's Republic of China
| |
Collapse
|
92
|
The Host Immune Response to Scedosporium/ Lomentospora. J Fungi (Basel) 2021; 7:jof7020075. [PMID: 33499053 PMCID: PMC7912657 DOI: 10.3390/jof7020075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Infections caused by the opportunistic pathogens Scedosporium/Lomentospora are on the rise. This causes problems in the clinic due to the difficulty in diagnosing and treating them. This review collates information published on immune response against these fungi, since an understanding of the mechanisms involved is of great interest in developing more effective strategies against them. Scedosporium/Lomentospora cell wall components, including peptidorhamnomannans (PRMs), α-glucans and glucosylceramides, are important immune response activators following their recognition by TLR2, TLR4 and Dectin-1 and through receptors that are yet unknown. After recognition, cytokine synthesis and antifungal activity of different phagocytes and epithelial cells is species-specific, highlighting the poor response by microglial cells against L. prolificans. Moreover, a great number of Scedosporium/Lomentospora antigens have been identified, most notably catalase, PRM and Hsp70 for their potential medical applicability. Against host immune response, these fungi contain evasion mechanisms, inducing host non-protective response, masking fungal molecular patterns, destructing host defense proteins and decreasing oxidative killing. In conclusion, although many advances have been made, many aspects remain to be elucidated and more research is necessary to shed light on the immune response to Scedosporium/Lomentospora.
Collapse
|
93
|
Pedan H, Janosova V, Hajtman A, Calkovsky V. Non-Reflex Defense Mechanisms of Upper Airway Mucosa: Possible Clinical Application. Physiol Res 2021; 69:S55-S67. [PMID: 32228012 DOI: 10.33549/physiolres.934404] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The sinonasal mucosa has an essential role in defense mechanisms of the upper respiratory tract. The innate immune system presents the primary defense against noxious microorganisms followed by induction of the adaptive immune mechanisms as a consequence of the presence of pathogens. This well-known activation of adaptive immune system in response to presence of the antigen on mucosal surfaces is now broadly applicated in vaccinology research. Prevention of infectious diseases belongs to substantial challenges in maintaining the population health. Non-invasive, easily applicable mucosal vaccination purposes various research opportunities that could be usable in daily practice. However, the existence of multiple limitations such as rapid clearance of vaccine from nasal mucosa by means of mucociliary transport represents a great challenge in development of safe and efficient vaccines. Here we give an updated view on nasal functions with focus on nasal mucosal immunity and its potential application in vaccination in nearly future.
Collapse
Affiliation(s)
- H Pedan
- Clinic of Otorhinolaryngology and Head and Neck Surgery, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, and Martin University Hospital, Martin, Slovak
| | | | | | | |
Collapse
|
94
|
Ghosh R, Bryant DL, Farone AL. Panax quinquefolius (North American Ginseng) Polysaccharides as Immunomodulators: Current Research Status and Future Directions. Molecules 2020; 25:E5854. [PMID: 33322293 PMCID: PMC7763949 DOI: 10.3390/molecules25245854] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Panax quinquefolius (North American ginseng, NAG) is a popular medicinal plant used widely in traditional medicine. NAG products are currently available in various forms such as roots, extracts, nutraceuticals, dietary supplements, energy drinks, etc. NAG polysaccharides are recognized as one of the major bioactive ingredients. However, most NAG reviews are focused on ginsenosides with little information on polysaccharides. NAG polysaccharides have demonstrated a therapeutic activity in numerous studies, in which many of the bioactivities involve regulation of the immune response. The purpose of this review is to summarize the structural features and the immunomodulatory properties of crude, partially purified, and pure polysaccharides isolated from NAG. Receptors of the innate immune system that potentially bind to NAG polysaccharides and the respective signal transduction pathways initiated by these compounds are discussed. Major challenges, recent innovations, and future directions in NAG polysaccharide research are also summarized.
Collapse
Affiliation(s)
- Rajarshi Ghosh
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA;
| | - Daniel L. Bryant
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Anthony L. Farone
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| |
Collapse
|
95
|
Jia Z, Wignall A, Prestidge C, Thierry B. An ex vivo investigation of the intestinal uptake and translocation of nanoparticles targeted to Peyer's patches microfold cells. Int J Pharm 2020; 594:120167. [PMID: 33309559 DOI: 10.1016/j.ijpharm.2020.120167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/19/2022]
Abstract
Diverse nanoparticulate systems have been engineered as vehicles towards enhancing the bioavailability of orally administrated vaccines. Substantial evidence suggests that targeting microfold cells (M cells) within Peyer's patches (PPs) is a prerequisite for vaccine-loaded nanocarriers to induce an effective antigen-specific immune response. Improved understanding of the contribution of M cells to sampling luminal nanoparticles into the underlying gut associated lymphoid tissues would accelerate the development of oral vaccine formulations. Herein, a novel clearing-based whole tissue mount/imaging technique was developed to enable the specific distribution of nanoparticles within ex vivo murine PPs to be quantitatively determined at the cellular level. This revealed that 200 nm nanoparticles modified with M cell targeting ligands (lectin Ulex europaeus agglutinin-1, UEA-1) were translocated into subepithelial domes 7.6 and 16.3 times greater than the non-targeted ones at 60 min and 120 min, respectively. This approach provides a new methodology to quantitatively investigate the transcytotic activity of M cells for particulate formulations, which may aid in the design of improved oral vaccines.
Collapse
Affiliation(s)
- Zhengyang Jia
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia
| | - Anthony Wignall
- UniSA Clinical and Health Science and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Clive Prestidge
- UniSA Clinical and Health Science and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, City West Campus, Adelaide, SA 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence Convergent Bio-Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Adelaide, SA 5095, Australia.
| |
Collapse
|
96
|
Mou MA, Keya NA, Islam M, Hossain MJ, Al Habib MS, Alam R, Rana S, Samad A, Ahammad F. Validation of CSN1S1 transcriptional expression, promoter methylation, and prognostic power in breast cancer using independent datasets. Biochem Biophys Rep 2020; 24:100867. [PMID: 33381666 PMCID: PMC7767798 DOI: 10.1016/j.bbrep.2020.100867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/16/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Breast cancer ranked second among most frequent cancer in the world playing a significant role in mortality rate. Having prior knowledge on differentially expressed genes in breast cell carcinoma elucidated important indications to understand the molecular mechanism underneath breast carcinogenesis. In this study we have investigated the distinguished CSN1S1 expression in human breast cancer. We have analyzed CSN1S1 mRNA expression between cancer and normal tissues using TCGA datasets. Moreover, analysis including promoter methylation, mutations, prognosis, co-expression, gene ontology, and pathways of CSN1S1 were performed by the TCGA Wanderer, UCSC Xena, cBioPortal, PrognoScan, UALCAN, and Enricher server. We have observed low mRNA expression and high promoter methylation of CSN1S1 in cancer tissues compared to normal tissues. Furthermore, we have also identified low mRNA expression in clinicopathological patients, as well as 9 deleterious mutations with highly co-expressed protein MRC1, and significantly related signaling pathways. We have found a positive correlation between the lower expression of CSN1S1 and patients surviving with breast cancer. Here we have concluded that CSN1S1 acts as a biomarker for the surveillance and prognosis of breast cancer, and also works as a novel therapeutic target at the molecular and pathway levels. Low transcriptional expression and low survival rate of CSN1S1 in breast cancer. The investigation of clinical profiles and mutational positions of CSN1S1 in breast cancer. The investigation of gene ontology and signaling pathway of CSN1S1 and their co-expressed genes. We identified CSN1S1 and also their co-expressed proteins are the potential biomarkers in breast cancer.
Collapse
Affiliation(s)
- Mohsina Akter Mou
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Bangladesh
| | - Nawshin Atia Keya
- Department of Microbiology, Noakhali Science and Technology University, Bangladesh
| | - Majharul Islam
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | | | - Md Syeed Al Habib
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Rahat Alam
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.,Laboraty of Computational Biology, Biological Solution Cantre (BiolSol Centre), Dhaka, Bangladesh
| | - Sohel Rana
- Department of Pharmacy, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Abdus Samad
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.,Laboraty of Computational Biology, Biological Solution Cantre (BiolSol Centre), Dhaka, Bangladesh
| | - Foysal Ahammad
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.,Laboraty of Computational Biology, Biological Solution Cantre (BiolSol Centre), Dhaka, Bangladesh.,Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, 21589, Saudi Arabia
| |
Collapse
|
97
|
Sadiq BA, Mantel I, Blander JM. A Comprehensive Experimental Guide to Studying Cross-Presentation in Dendritic Cells In Vitro. CURRENT PROTOCOLS IN IMMUNOLOGY 2020; 131:e115. [PMID: 33316130 PMCID: PMC9060150 DOI: 10.1002/cpim.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cross-presentation was first observed serendipitously in the 1970s. The importance of it was quickly realized and subsequently attracted great attention from immunologists. Since then, our knowledge of the ability of certain antigen presenting cells to internalize, process, and load exogenous antigens onto MHC-I molecules to cross-prime CD8+ T cells has increased significantly. Dendritic cells (DCs) are exceptional cross-presenters, thus making them a great tool to study cross-presentation but the relative rarity of DCs in circulation and in tissues makes it challenging to isolate sufficient numbers of cells to study this process in vitro. In this paper, we describe in detail two methods to culture DCs from bone-marrow progenitors and a method to expand the numbers of DCs present in vivo as a source of endogenous bona-fide cross-presenting DCs. We also describe methods to assess cross-presentation by DCs using the activation of primary CD8+ T cells as a readout. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Isolation of bone marrow progenitor cells Basic Protocol 2: In vitro differentiation of dendritic cells with GM-CSF Support Protocol 1: Preparation of conditioned medium from GM-CSF producing J558L cells Basic Protocol 3: In vitro differentiation of dendritic cells with Flt3L Support Protocol 2: Preparation of Flt3L containing medium from B16-Flt3L cells Basic Protocol 4: Expansion of cDC1s in vivo for use in ex vivo experiments Basic Protocol 5: Characterizing resting and activated dendritic cells Basic Protocol 6: Dendritic cell stimulation, antigenic cargo, and fixation Support Protocol 3: Preparation of model antigen coated microbeads Support Protocol 4: Preparation of apoptotic cells Support Protocol 5: Preparation of recombinant bacteria Basic Protocol 7: Immunocytochemistry immunofluorescence (ICC/IF) Support Protocol 6: Preparation of Alcian blue-coated coverslips Basic Protocol 8: CD8+ T cell activation to assess cross-presentation Support Protocol 7: Isolation and labeling of CD8+ T cells with CFSE.
Collapse
Affiliation(s)
- Barzan A. Sadiq
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
| | - Ian Mantel
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, New York
| | - J. Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, New York
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, New York
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, New York
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
98
|
Zhou K, Huang Y, Chen Z, Du X, Qin J, Wen L, Ma H, Pan X, Lin Y. Liver and spleen transcriptome reveals that Oreochromis aureus under long-term salinity stress may cause excessive energy consumption and immune response. FISH & SHELLFISH IMMUNOLOGY 2020; 107:469-479. [PMID: 33181338 DOI: 10.1016/j.fsi.2020.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/14/2020] [Accepted: 11/09/2020] [Indexed: 06/11/2023]
Abstract
To investigate the physiological responses of Oreochromis aureus to salinity fluctuations at the molecular level. We used RNA-seq to explore the differentially expressed genes (DEGs) in the liver and spleen of O. aureus at 0, 3, 7 and 11 ppt (parts per thousand) salinity levels. Herein, De novo assembly generated 71,009 O. aureus unigenes, of which 34,607 were successfully mapped to the four major databases. A total of 120 shared DEGs were identified in liver and spleen transcripts, of which 83 were up-regulated and 37 were down-regulated. GO and KEGG analysis found a total of 26 significant pathways, mainly including energy metabolism, immune response, ion transporters and signal transduction. The trend module category of DEGs showed that the genes (e.g., FASN, ODC1, CD22, MRC, TRAV and SLC7 family) involved in the change-stable-change (1) and the constant-change categories (2) were highly sensitive to salinity fluctuations, which were of great value for further study. Based on these results, it would help provide basic data for fish salinity acclimation, and provide new insights into evolutionary response of fish to various aquatic environments in the long-term stress adaptation mechanism.
Collapse
Affiliation(s)
- Kangqi Zhou
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Yin Huang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Zhong Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Xuesong Du
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Junqi Qin
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Luting Wen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Huawei Ma
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Xianhui Pan
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China.
| | - Yong Lin
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, 530021, China.
| |
Collapse
|
99
|
Increased Levels of Soluble CD206 Associated with Rapidly Progressive Interstitial Lung Disease in Patients with Dermatomyositis. Mediators Inflamm 2020; 2020:7948095. [PMID: 33192174 PMCID: PMC7641712 DOI: 10.1155/2020/7948095] [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: 05/26/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 01/02/2023] Open
Abstract
Objective Soluble CD206 (sCD206) is considered a macrophage activation marker, and a previous study proved it as a potential biomarker to predict the severity of anti-melanoma differentiation-associated gene 5- (anti-MDA-5-) positive dermatomyositis- (DM-) associated interstitial lung disease (ILD). To investigate the role of sCD206 in various subtypes of DM, we evaluated the serum level of sCD206 in patients with different myositis-specific autoantibodies besides anti-MDA-5 and clarified its clinical significance. Methods Commercial enzyme-linked immunosorbent assay kits were used to detect serum concentrations of sCD206 in 150 patients with DM and 52 healthy controls (HCs). Correlations between sCD206 levels and clinical features, laboratory examinations, and pulmonary function test parameters were analysed. Results The median concentrations of serum sCD206 in DM patients were significantly higher than those in HCs (p < 0.0001). Furthermore, median sCD206 levels were elevated in patients with ILD (p = 0.001), especially in those with rapidly progressive ILD (RP-ILD) (p < 0.0001). In addition, sCD206 levels were negatively correlated with the pulmonary function test results, including the percent predicted forced vital capacity (r = −0.234, p = 0.023), percent predicted forced expiratory volume in one second (r = −0.225, p = 0.030), and percent predicted carbon monoxide diffusion capacity (r = −0.261, p = 0.014). Age- and gender-adjusted multivariable analysis showed that sCD206 was an independent prognostic factor for RP-ILD in patients with DM. A longitudinal study showed that sCD206 levels were positively correlated with the physician global assessment visual analog scale scores (β = 54.201, p = 0.001). Conclusion Serum sCD206 levels were significantly increased in patients with DM and significantly associated with RP-ILD, suggesting that sCD206 is an important biological predictor of RP-ILD in patients with DM.
Collapse
|
100
|
Gula G, Rumiński S, Niderla-Bielińska J, Jasińska A, Kiernozek E, Jankowska-Steifer E, Flaht-Zabost A, Ratajska A. Potential functions of embryonic cardiac macrophages in angiogenesis, lymphangiogenesis and extracellular matrix remodeling. Histochem Cell Biol 2020; 155:117-132. [PMID: 33130914 PMCID: PMC7847984 DOI: 10.1007/s00418-020-01934-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
The role of cardiac tissue macrophages (cTMs) during pre- and postnatal developmental stages remains in many aspects unknown. We aimed to characterize cTM populations and their potential functions based on surface markers. Our in situ studies of immunostained cardiac tissue specimens of murine fetuses (from E11to E17) revealed that a significant number of embryonic cTMs (phenotyped by CD45, CD68, CD64, F4/80, CD11b, CD206, Lyve-1) resided mostly in the subepicardial space, not in the entire myocardial wall, as observed in adult individuals. cTMs accompanied newly developed blood and lymphatic vessels adhering to vessel walls by cellular processes. A subpopulation of CD68-positive cells was found to form accumulations in areas of massive apoptosis during the outflow tract remodeling and shortening. Flow cytometry analysis at E14 and E17 stages revealed newly defined three subpopulations:CD64low, CD64highCD206-and CD64highCD206+. The levels of mRNA expression for genes related to regulation of angiogenesis (VEGFa, VEGFb, VEGFc, bFGF), lymphangiogenesis (VEGFc) and extracellular matrix (ECM) remodeling (MMP13, Arg1, Ym1/Chil3, Retlna/FIZZ1) differed among the selected populations and/or embryonic stages. Our results demonstrate a diversity of embryonic cTMs and their tissue-specific locations, suggesting their various potential roles in regulating angiogenesis, lymphangiogenesis and ECM remodeling.
Collapse
Affiliation(s)
- Grzegorz Gula
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of Ophthalmology, Central Clinical Hospital of the MSWiA, Warsaw, Poland
| | - Sławomir Rumiński
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland.,Centre for Preclinical Research and Technology, Warsaw, Poland
| | | | - Agnieszka Jasińska
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Student's Scientific Circle at Department of Pathology MUW, Warsaw, Poland
| | | | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Flaht-Zabost
- Department of Pathology, Medical University of Warsaw, Chałubińskiego 5, 02-004, Warsaw, Poland
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Chałubińskiego 5, 02-004, Warsaw, Poland.
| |
Collapse
|