1
|
Ji M, Liu H, Wei M, Shi D, Gou J, Yin T, He H, Tang X, Chen C, Zhang Y. Redox-sensitive disulfide-bridged self-assembled nanoparticles of dexamethasone with high drug loading for acute lung injury therapy. Int J Pharm 2024; 664:124600. [PMID: 39159858 DOI: 10.1016/j.ijpharm.2024.124600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Acute lung injury (ALI) arises from an excessive inflammatory response, usually progressing to acute respiratory distress syndrome (ARDS) if not promptly addressed. There is currently a limited array of effective treatments available for ALI. In this study, we developed disulfide bond-bridged prodrug self-assembled nanoparticles (referred to as DSSS NPs). These nanoparticles were consisted of Dexamethasone (Dex) and stearic acid (SA), and were designed to target and treat ALI. DSSS NPs demonstrated a substantial drug loading capacity with 37.75 % of Dex, which is much higher than conventional nanomedicines (usually < 10 %). Moreover, they exhibited the potential to specifically target injured lung tissue and inflammatory microenvironment-responsive release drugs. Consequently, DSSS NPs reduced significantly the levels of pro-inflammatory cytokines and tissue damage in mice with ALI induced by lipopolysaccharide (LPS). Overall, DSSS NPs offer a promising strategy for treatment of acute lung injury.
Collapse
Affiliation(s)
- Muse Ji
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Hongbing Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Mingli Wei
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Dongmei Shi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Tian Yin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China.
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China
| | - Chengjun Chen
- Beijing Nuokangda Pharmaceutical Co., Ltd, Beijing, 100176 Beijing, China.
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning, China.
| |
Collapse
|
2
|
Corrêa-Junior D, Bastos de Andrade I, Alves V, Avellar-Moura I, Brito de Souza Rabello V, Valdez AF, Nimrichter L, Zancopé-Oliveira RM, Ribeiro de Sousa Araújo G, Almeida-Paes R, Frases S. Unveiling the Morphostructural Plasticity of Zoonotic Sporotrichosis Fungal Strains: Possible Implications for Sporothrix brasiliensis Virulence and Pathogenicity. J Fungi (Basel) 2023; 9:701. [PMID: 37504690 PMCID: PMC10381685 DOI: 10.3390/jof9070701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Sporotrichosis is a fungal infection caused by Sporothrix species, with Sporothrix brasiliensis as a prevalent pathogen in Latin America. Despite its clinical importance, the virulence factors of S. brasiliensis and their impact on the pathogenesis of sporotrichosis are still poorly understood. This study evaluated the morphostructural plasticity of S. brasiliensis, a fungus that causes sporotrichosis. Three cell surface characteristics, namely cell surface hydrophobicity, Zeta potential, and conductance, were assessed. Biofilm formation was also analyzed, with measurements taken for biomass, extracellular matrix, and metabolic activity. In addition, other potential and poorly studied characteristics correlated with virulence such as lipid bodies, chitin, and cell size were evaluated. The results revealed that the major phenotsypic features associated with fungal virulence in the studied S. brasiliensis strains were chitin, lipid bodies, and conductance. The dendrogram clustered the strains based on their overall similarity in the production of these factors. Correlation analyses showed that hydrophobicity was strongly linked to the production of biomass and extracellular matrix, while there was a weaker association between Zeta potential and size, and lipid bodies and chitin. This study provides valuable insights into the virulence factors of S. brasiliensis and their potential role in the pathogenesis of sporotrichosis.
Collapse
Affiliation(s)
- Dario Corrêa-Junior
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Iara Bastos de Andrade
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Vinicius Alves
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Igor Avellar-Moura
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Vanessa Brito de Souza Rabello
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Alessandro Fernandes Valdez
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21040-900, Brazil
| | - Leonardo Nimrichter
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21040-900, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Rosely Maria Zancopé-Oliveira
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Glauber Ribeiro de Sousa Araújo
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Rodrigo Almeida-Paes
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| | - Susana Frases
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Rede Micologia RJ, FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Lobognon VD, Alard JE. Could AMPs and B-cells be the missing link in understanding periodontitis? Front Immunol 2022; 13:887147. [PMID: 36211356 PMCID: PMC9532695 DOI: 10.3389/fimmu.2022.887147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/01/2022] [Indexed: 12/29/2022] Open
Abstract
Periodontal diseases are common inflammatory conditions characterized by bone loss in response to simultaneous bacterial aggression and host defenses. The etiology of such diseases is still not completely understood, however. It has been shown that specific pathogens involved in the build-up of dysbiotic biofilms participate actively in the establishment of periodontitis. This multifactorial pathology also depends on environmental factors and host characteristics, especially defenses. The immune response to the pathogens seems to be critical in preventing the disease from starting but also contributes to tissue damage. It is known that small molecules known as antimicrobial peptides (AMPs) are key actors in the innate immune response. They not only target microbes, but also act as immuno-modulators. They can help to recruit or activate cells such as neutrophils, monocytes, dendritic cells, or lymphocytes. AMPs have already been described in the periodontium, and their expression seems to be connected to disease activity. Alpha and beta defensins and LL37 are the AMPs most frequently linked to periodontitis. Additionally, leukocyte infiltrates, especially B-cells, have also been linked to the severity of periodontitis. Indeed, the particular subpopulations of B-cells in these infiltrates have been linked to inflammation and bone resorption. A link between B-cells and AMP could be relevant to understanding B-cells' action. Some AMP receptors, such as chemokines receptors, toll-like receptors, or purinergic receptors, have been shown to be expressed by B-cells. Consequently, the action of AMPs on B-cell subpopulations could participate to B-cell recruitment, their differentiation, and their implication in both periodontal defense and destruction.
Collapse
Affiliation(s)
- Vanessa Dominique Lobognon
- B lymphocytes, Autoimmunity and Immunotherapies (LBAI), Mixed Research Unit (UMR)1227 INSERM, University of Brest, Brest, France
| | - Jean-Eric Alard
- B lymphocytes, Autoimmunity and Immunotherapies (LBAI), Mixed Research Unit (UMR)1227 INSERM, University of Brest, Brest, France,Service d’Odontologie, University Hospital (CHU) de Brest, Brest, France,*Correspondence: Jean-Eric Alard,
| |
Collapse
|
4
|
Fouda MA, Mohamed YF, Fernandez R, Ruben PC. Anti-inflammatory effects of cannabidiol against lipopolysaccharides in cardiac sodium channels. Br J Pharmacol 2022; 179:5259-5272. [PMID: 35906756 DOI: 10.1111/bph.15936] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/13/2022] [Accepted: 07/24/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Sepsis, caused by a dysregulated host response to infections, can lead to cardiac arrhythmias. However, the mechanisms underlying sepsis-induced inflammation, and how inflammation provokes cardiac arrhythmias, are not well understood. We hypothesized that CBD may ameliorate lipopolysaccharides (LPS)-induced cardiotoxicity via Toll-like receptor 4 (TLR-4) and cardiac sodium channels (Nav1.5). METHODS AND RESULTS We incubated human immune cells (THP-1 macrophages) with LPS for 24 hours, then extracted the THP-1 incubation media. ELISA assay showed that LPS (1 or 5 μg/ml), in a concentration-dependent manner, or MPLA (TLR-4 agonist, 5 μg/ml) stimulated the THP-1 cells to release inflammatory cytokines (TNF-α and IL-6). Prior incubation (4 hours) with cannabidiol (CBD: 5 μM) or C34 (TLR-4 antagonist: 5 μg/ml) inhibited LPS and MPLA-induced release of both IL-6 and TNF-α. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) were subsequently incubated for 24 hours in the media extracted from THP-1 cells incubated with LPS, MPLA alone, or in combination with CBD or C34. Voltage-clamp experiments showed a right shift in the voltage dependence of Nav1.5 activation, steady state fast inactivation (SSFI), increased persistent current and prolonged in silico action potential duration in hiSPC-CM incubated in the LPS or MPLA-THP-1 media. Co-incubation with CBD or C34 rescued the biophysical dysfunction caused by LPS and MPLA. CONCLUSION Our results suggest that CBD may protect against sepsis-induced inflammation and subsequent arrhythmias through (i) inhibition of the release of inflammatory cytokines, antioxidant and anti-apoptotic effects and/or (ii) direct effect on Nav1.5.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Yasmine Fathy Mohamed
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada.,Department of Microbiology and Immunology, Alexandria University, Alexandria, Egypt
| | - Rachel Fernandez
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
5
|
Choi J, Ki CS. Ultrasonication, immune activity, and photocrosslinked microgel formation of pectic polysaccharide isolated from root bark of Ulmus davidiana var. japonica (Rehder) Nakai. Int J Biol Macromol 2022; 211:535-544. [PMID: 35569684 DOI: 10.1016/j.ijbiomac.2022.05.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022]
Abstract
The root bark of Ulmus davidiana var. japonica (Rehder) Nakai (Japanese elm) has been used for inflammatory disease treatments. In this work, we isolated pectic polysaccharides from the root bark of U. davidiana (UDP) and explored the immune activities of intact and ultrasonicated UDP on human macrophages. The UDP-treated macrophages showed a proinflammatory response, indicating classical activation via Toll-like receptor-mediated recognition. For hydrogel formation, the ultrasonicated UDP was modified with methacrylate groups, then subjected to photocrosslinking. The formed bulk hydrogel was pulverized into microgels by homogenization, and the microgel size was modulated for macrophage phagocytosis. The UDP microgel-treated macrophages displayed microgel internalization and classical activation that involved upregulation of M1 polarization markers (IL6, TNF-α, and CCR7), indicating that the microgel can be used as a carrier for macrophage-targeted drug delivery.
Collapse
Affiliation(s)
- Jaeho Choi
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul 08826, Republic of Korea
| | - Chang Seok Ki
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
6
|
TLR22-mediated activation of TNF-α-caspase-1/IL-1β inflammatory axis leads to apoptosis of Aeromonas hydrophila-infected macrophages. Mol Immunol 2021; 137:114-123. [PMID: 34242920 DOI: 10.1016/j.molimm.2021.06.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/28/2021] [Accepted: 06/29/2021] [Indexed: 02/08/2023]
Abstract
Toll-like receptors (TLRs) represent first line of host defence against microbes. Amongst different TLRs, TLR22 is exclusively expressed in non-mammalian vertebrates, including fish. The precise role of TLR22 in fish-immunity remains abstruse. Herein, we used headkidney macrophages (HKM) from Clarias gariepinus and deciphered its role in fish-immunity. Highest tlr22 expression was observed in the immunocompetent organ - headkidney; nonetheless expression in other tissues suggests its possible involvement in non-immune sites also. Aeromonas hydrophila infection up-regulates tlr22 expression in HKM. Our RNAi based study suggested TLR22 restricts intracellular survival of A. hydrophila. Inhibitor and RNAi studies further implicated TLR22 induces pro-inflammatory cytokines TNF-α and IL-1β. We observed heightened caspase-1 activity and our results suggest the role of TLR22 in activating TNF-α/caspase-1/IL-1β cascade leading to caspase-3 mediated apoptosis of A. hydrophila-infected HKM. We conclude, TLR22 plays critical role in immune-surveillance and triggers pro-inflammatory cytokines leading to caspase mediated HKM apoptosis and pathogen clearance.
Collapse
|
7
|
Muñoz-Caro T, Gibson AJ, Conejeros I, Werling D, Taubert A, Hermosilla C. The Role of TLR2 and TLR4 in Recognition and Uptake of the Apicomplexan Parasite Eimeria bovis and Their Effects on NET Formation. Pathogens 2021; 10:pathogens10020118. [PMID: 33498871 PMCID: PMC7912269 DOI: 10.3390/pathogens10020118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Bovine polymorphonuclear neutrophils (PMN) constitutively express the Toll-like receptors (TLRs) TLR2 and TLR4 and have been shown to generate Neutrophil extracellular traps (NETs) upon exposure to Eimeria bovis. The present work investigated the role of TLR2 and TLR4 in the recognition and uptake of E. bovis sporozoites, IL-8 production and neutrophil extracellular trap (NET) formation. METHODS TLR expression was performed by flow cytometric analysis on PMN exposed to live carboxyfluorescein succinimidyl ester (CFSE)-stained sporozoites. Supernatants of PMN exposed to different E. bovis sporozoite preparations and antigens in the absence or presence of TLR antibodies were assessed for IL-8 secretion. Cells were exposed to sporozoite preparations and assessed for the activation of transcription factor NF-κB using a luciferase reporter assay. Immunofluorescence analysis was done to investigate TLR2 and TLR4 surface expression and NET formation on bovine PMN exposed to vital sporozoites. RESULTS we observed significantly increased TLR2 and TLR4 expression with a mean increase in expression that was greater for TLR2 than TLR4. This upregulation neither inhibited nor promoted sporozoite phagocytosis by bovine PMN. Live sporozoites together with anti-TLR2 mAb resulted in a significant enhancement of IL-8 production. NF-κB activation was more strongly induced in TLR2-HEK cells than in TLR4/MD2-HEK cells exposed to heat-killed sporozoites and antigens. Immunofluorescence analysis showed TLR-positive signals on the surface of PMN and concomitant NET formation. CONCLUSIONS This is the first report on E. bovis-induced concomitant TLR2 and TLR4 expression during bovine PMN-derived NETosis.
Collapse
Affiliation(s)
- Tamara Muñoz-Caro
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (T.M.-C.); (I.C.); (A.T.)
- Escuela de Medicina Veterinaria, Facultad de Recursos Naturales y Medicina Veterinaria, Universidad Santo Tomás, Talca 3460000, Chile
| | - Amanda J. Gibson
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK; (A.J.G.); (D.W.)
- Centre of Excellence in Bovine Tuberculosis, Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Wales SY23 3FD, UK
| | - Iván Conejeros
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (T.M.-C.); (I.C.); (A.T.)
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK; (A.J.G.); (D.W.)
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (T.M.-C.); (I.C.); (A.T.)
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (T.M.-C.); (I.C.); (A.T.)
- Correspondence:
| |
Collapse
|
8
|
A Fish Leukocyte Immune-Type Receptor Uses a Novel Intracytoplasmic Tail Networking Mechanism to Cross-Inhibit the Phagocytic Response. Int J Mol Sci 2020; 21:ijms21145146. [PMID: 32708174 PMCID: PMC7404264 DOI: 10.3390/ijms21145146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/04/2023] Open
Abstract
Channel catfish (Ictalurus punctatus) leukocyte immune-type receptors (IpLITRs) are a family of immunoregulatory proteins shown to regulate several innate immune cell effector responses, including phagocytosis. The precise mechanisms of IpLITR-mediated regulation of the phagocytic process are not entirely understood, but we have previously shown that different IpLITR-types use classical as well as novel pathways for controlling immune cell-mediated target engulfment. To date, all functional assessments of IpLITR-mediated regulatory actions have focused on the independent characterization of select IpLITR-types in transfected cells. As members of the immunoglobulin superfamily, many IpLITRs share similar extracellular Ig-like domains, thus it is possible that various IpLITR actions are influenced by cross-talk mechanisms between different IpLITR-types; analogous to the paired innate receptor paradigm in mammals. Here, we describe in detail the co-expression of different IpLITR-types in the human embryonic AD293 cell line and examination of their receptor cross-talk mechanisms during the regulation of the phagocytic response using imaging flow cytometry, confocal microscopy, and immunoprecipitation protocols. Overall, our data provides interesting new insights into the integrated control of phagocytosis via the antagonistic networking of independent IpLITR-types that requires the selective recruitment of inhibitory signaling molecules for the initiation and sustained cross-inhibition of phagocytosis.
Collapse
|
9
|
Abstract
Phagocytosis is a complex process by which cells within most organ systems remove pathogens and cell debris. Phagocytosis is usually followed by inflammatory pathway activation, which promotes pathogen elimination and inhibits pathogen growth. Delayed pathogen elimination is the first step in sepsis development and a key factor in sepsis resolution. Phagocytosis thus has an important role during sepsis and likely contributes to all of its clinical stages. However, only a few studies have specifically explored and characterized phagocytic activity during sepsis. Here, we describe the phagocytic processes that occur as part of the immune response preceding sepsis onset and identify the elements of phagocytosis that might constitute a predictive marker of sepsis outcomes. First, we detail the key features of phagocytosis, including the main receptors and signaling hallmarks associated with different phagocytic processes. We then discuss how the initial events of phagosome formation and cytoskeletal remodeling might be associated with known sepsis features, such as a cytokine-driven hyperinflammatory response and immunosuppression. Finally, we highlight the unresolved mechanisms of sepsis development and progression and the need for cross-disciplinary approaches to link the clinical complexity of the disease with basic cellular and molecular mechanisms.
Collapse
|
10
|
Uher O, Caisova V, Hansen P, Kopecky J, Chmelar J, Zhuang Z, Zenka J, Pacak K. Coley's immunotherapy revived: Innate immunity as a link in priming cancer cells for an attack by adaptive immunity. Semin Oncol 2019; 46:385-392. [PMID: 31739997 DOI: 10.1053/j.seminoncol.2019.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
Abstract
There is no doubt that immunotherapy lies in the spotlight of current cancer research and clinical trials. However, there are still limitations in the treatment response in certain types of tumors largely due to the presence of the complex network of immunomodulatory and immunosuppressive pathways. These limitations are not likely to be overcome by current immunotherapeutic options, which often target isolated steps in immune pathways preferentially involved in adaptive immunity. Recently, we have developed an innovative anti-cancer immunotherapeutic strategy that initially elicits a strong innate immune response with subsequent activation of adaptive immunity in mouse models. Robust primary innate immune response against tumor cells is induced by toll-like receptor ligands and anti-CD40 agonistic antibodies combined with the phagocytosis-stimulating ligand mannan, anchored to a tumor cell membrane by biocompatible anchor for membrane. This immunotherapeutic approach results in a dramatic therapeutic response in large established murine subcutaneous tumors including melanoma, sarcoma, pancreatic adenocarcinoma, and pheochromocytoma. Additionally, eradication of metastases and/or long-lasting resistance to subsequent re-challenge with tumor cells was also accomplished. Current and future advantages of this immunotherapeutic approach and its possible combinations with other available therapies are discussed in this review.
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA; Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Per Hansen
- Immunoaction LLC, Charlotte, Vermont, VT 05445, USA
| | - Jan Kopecky
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Jindrich Chmelar
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA.
| |
Collapse
|
11
|
Nawab A, An L, Wu J, Li G, Liu W, Zhao Y, Wu Q, Xiao M. Chicken toll-like receptors and their significance in immune response and disease resistance. Int Rev Immunol 2019; 38:284-306. [PMID: 31662000 DOI: 10.1080/08830185.2019.1659258] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Infectious diseases are a major challenge for the poultry industry that causes widespread production losses. Thus, management and control of poultry health and diseases are essential for the viability of the industry. Toll-like receptors are best characterized as membrane-bound receptors that perform a central role in immune homeostasis and disease resistance by recognition of pathogen-associated molecular patterns. In response to pathogen recognition, TLRs initiate both innate and adaptive immune responses which may help to develop immunomodulatory therapeutics for TLR associated diseases. Vaccination produces specific immunity in the animal's body towards pathogens. However, due to certain disadvantages of vaccines, (inactivation of attenuated pathogens into the virulent strains and weak immunogenicity of inactivated vaccines) there is a crucial need to develop the safe and effective therapeutic intervention. TLR ligands have been classified as a potential adjuvant against the infectious diseases in farm animals. TLR adjuvants induce both specific and nonspecific immune responses in chickens to combat several bacterial, viral and parasitic diseases. Therefore, the aim of this review was to explore the chicken TLR4 and their role in immune responses and disease resistance to develop disease resistance poultry breeds in future.
Collapse
Affiliation(s)
- Aamir Nawab
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China.,Faculty of Veterinary Medicine, PMAS- Arid Agriculture University Rawalpindi, Rawalpindi, Pakistan
| | - Lilong An
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiang Wu
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Guanghui Li
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Wenchao Liu
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yi Zhao
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Qimin Wu
- Mechanical and Power Engineering College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Mei Xiao
- Department of Animal Science, Guangdong Ocean University, Zhanjiang, Guangdong, China
| |
Collapse
|
12
|
Lv Z, Wang L, Jia Z, Sun J, Wang W, Liu Z, Qiu L, Wang M, Song L. Hemolymph C1qDC promotes the phagocytosis of oyster Crassostrea gigas hemocytes by interacting with the membrane receptor β-integrin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 98:42-53. [PMID: 30995452 DOI: 10.1016/j.dci.2019.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 06/09/2023]
Abstract
Phagocytosis constitutes a conserved cellular process for multicellular animals to ingest or engulf other cells or particles, which is facilitated by the use of opsonins to bind foreign particles and interact with cell surface receptors. The invertebrate secreted C1q domain-containing proteins (C1qDCs) have been reported to exhibit opsonic activity, while the detailed mechanisms of opsonization still remain unclear. In the present study, a C1qDC (designated as CgC1qDC-5) with opsonic activity was identified from the hemolymph of oyster Crassostrea gigas. CgC1qDC-5 exhibited the ability to bind pathogen-associated molecular patterns (PAMPs) of lipopolysaccharides (LPS) and Lipid A. It could also bind and agglutinate Gram-negative bacteria Escherichia coli, Vibrio splendidus and Vibrio anguillarum, whereas the agglutinating activity could be inhibited by LPS. In addition, CgC1qDC-5 could enhance the phagocytosis of hemocytes toward E. coli, V. splendidus, and V. anguillarum. GST pull-down and surface plasmon resonance assays in vitro revealed that CgC1qDC-5 could interact with β-integrin (CgIntegrin). In vivo, CgC1qDC-5 was observed to bind hemocytes and co-localized with CgIntegrin on the cell membrane of hemocytes. Antibody-mediated blockage of CgIntegrin hindered the CgC1qDC-5-enhanced hemocytic phagocytosis. CgIntegrin also exhibited the ability to bind the Gram-negative bacteria E. coli, V. splendidus, V. anguillarum and Vibrio parahaemolyticus, and PAMP of LPS, but not Lipid A. A phagocytosis assay demonstrated that CgIntegrin could directly mediate phagocytosis toward bacteria as a phagocytic receptor. These results collectively suggested that CgC1qDC-5 could serve as an opsonin to recognize and bind bacteria, and subsequently interact with CgIntegrin on the hemocyte surface to enhance the CgIntegrin-mediated phagocytosis in oyster.
Collapse
Affiliation(s)
- Zhao Lv
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingling Wang
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Zhihao Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Zhaoqun Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Linsheng Song
- Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
13
|
Kumar R, Kumar P. Yeast-based vaccines: New perspective in vaccine development and application. FEMS Yeast Res 2019; 19:5298404. [PMID: 30668686 DOI: 10.1093/femsyr/foz007] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
In presently licensed vaccines, killed or attenuated organisms act as a source of immunogens except for peptide-based vaccines. These conventional vaccines required a mass culture of associated or related organisms and long incubation periods. Special requirements during storage and transportation further adds to the cost of vaccine preparations. Availability of complete genome sequence, well-established genetic, inherent natural adjuvant and non-pathogenic nature of yeast species viz. Saccharomyces cerevisiae, Pichia pastoris makes them an ideal model system for the development of vaccines both for public health and for on-farm consumption. In this review, we compile the work in this emerging field during last two decades with major emphases on S. cerevisiae and P. pastoris which are routinely used worldwide for expression of heterologous proteins with therapeutic value against infectious diseases along with possible use in cancer therapy. We also pointed towards the developments in use of whole recombinant yeast, yeast surface display and virus-like particles as a novel strategy in the fight against infectious diseases and cancer along with other aspects including suitability of yeast in vaccines preparations, yeast cell wall component as an immune stimulator or modulator and present status of yeast-based vaccines in clinical trials.
Collapse
Affiliation(s)
- Ravinder Kumar
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Piyush Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400 076, Maharashtra, India
| |
Collapse
|
14
|
Kim TH, Lee HC, Kim JH, Hewawaduge CY, Chathuranga K, Chathuranga WAG, Ekanayaka P, Wijerathne HMSM, Kim CJ, Kim E, Lee JS. Fas-associated factor 1 mediates NADPH oxidase-induced reactive oxygen species production and proinflammatory responses in macrophages against Listeria infection. PLoS Pathog 2019; 15:e1008004. [PMID: 31412082 PMCID: PMC6709923 DOI: 10.1371/journal.ppat.1008004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/26/2019] [Accepted: 07/27/2019] [Indexed: 11/29/2022] Open
Abstract
Fas-associated factor 1 is a death-promoting protein that induces apoptosis by interacting with the Fas receptor. Until now, FAF1 was reported to interact potentially with diverse proteins and to function as a negative and/or positive regulator of several cellular possesses. However, the role of FAF1 in defense against bacterial infection remains unclear. Here, we show that FAF1 plays a pivotal role in activating NADPH oxidase in macrophages during Listeria monocytogenes infection. Upon infection by L. monocytogenes, FAF1 interacts with p67phox (an activator of the NADPH oxidase complex), thereby facilitating its stabilization and increasing the activity of NADPH oxidase. Consequently, knockdown or ectopic expression of FAF1 had a marked effect on production of ROS, proinflammatory cytokines, and antibacterial activity, in macrophages upon stimulation of TLR2 or after infection with L. monocytogenes. Consistent with this, FAF1gt/gt mice, which are knocked down in FAF1, showed weaker inflammatory responses than wild-type mice; these weaker responses led to increased replication of L. monocytogenes. Collectively, these findings suggest that FAF1 positively regulates NADPH oxidase-mediated ROS production and antibacterial defenses. Phagocytic NADPH oxidase plays a pivotal role in generating reactive oxygen species (ROS) and in defense against bacterial infections such as L. monocytogenes. ROS eliminate phagocytosed bacteria directly and are implicated in transduction of signals that mediate inflammatory responses. Here, we show that the apoptotic protein FAF1 regulates ROS production in macrophages by regulating phagocytic NADPH oxidase activity upon infection by L. monocytogenes. FAF1 interacts directly with and stabilizes p67phox, a regulatory protein of the phagocytic NADPH oxidase complex, to induce ROS production during L. monocytogenes infection. Production of ROS leads to release of proinflammatory cytokines, chemokines and, ultimately, to bacterial clearance. Interestingly, FAF1gt/gt mice deficient in FAF1 expression exhibit weakened inflammatory responses and are thus more vulnerable to bacterial infection than FAF1+/+ mice. This study reveals that FAF1 is a crucial regulator that induces inflammatory responses to bacterial infection via ROS production.
Collapse
Affiliation(s)
- Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Cheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jae-Hoon Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - C. Y. Hewawaduge
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | | | - Pathum Ekanayaka
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - H. M. S. M. Wijerathne
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eunhee Kim
- College of Biological Sciences and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Awasthi S, Singh B, Ramani V, Xie J, Kosanke S. TLR4-interacting SPA4 peptide improves host defense and alleviates tissue injury in a mouse model of Pseudomonas aeruginosa lung infection. PLoS One 2019; 14:e0210979. [PMID: 30689633 PMCID: PMC6349318 DOI: 10.1371/journal.pone.0210979] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/04/2019] [Indexed: 11/18/2022] Open
Abstract
Interaction between surfactant protein-A (SP-A) and toll-like receptor (TLR)4 plays a critical role in host defense. In this work, we studied the host defense function of SPA4 peptide (amino acids GDFRYSDGTPVNYTNWYRGE), derived from the TLR4-interacting region of SP-A, against Pseudomonas aeruginosa. We determined the binding of SPA4 peptide to live bacteria, and its direct antibacterial activity against P. aeruginosa. Pro-phagocytic and anti-inflammatory effects were investigated in JAWS II dendritic cells and primary alveolar macrophages. The biological relevance of SPA4 peptide was evaluated in a mouse model of acute lung infection induced by intratracheal challenge with P. aeruginosa. Our results demonstrate that the SPA4 peptide does not interact with or kill P. aeruginosa when cultured outside the host. The SPA4 peptide treatment induces the uptake and localization of bacteria in the phagolysosomes of immune cells. At the same time, the secreted amounts of TNF-α are significantly reduced in cell-free supernatants of SPA4 peptide-treated cells. In cells overexpressing TLR4, the TLR4-induced phagocytic response is maintained, but the levels of TLR4-stimulated TNF-α are reduced. Furthermore, our results demonstrate that the therapeutic administration of SPA4 peptide reduces bacterial burden, inflammatory cytokines and chemokines, intracellular signaling, and lactate levels, and alleviates lung edema and tissue damage in P. aeruginosa-infected mice. Together, our results suggest that the treatment with SPA4 peptide can help control the bacterial burden, inflammation, and tissue injury in a P. aeruginosa lung infection model.
Collapse
Affiliation(s)
- Shanjana Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States of America
- * E-mail:
| | - Bhupinder Singh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States of America
| | - Vijay Ramani
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States of America
| | - Jun Xie
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, United States of America
| | - Stanley Kosanke
- Department of Pathology, OUHSC, Oklahoma City, Oklahoma, United States of America
| |
Collapse
|
16
|
Gowda DC, Wu X. Parasite Recognition and Signaling Mechanisms in Innate Immune Responses to Malaria. Front Immunol 2018; 9:3006. [PMID: 30619355 PMCID: PMC6305727 DOI: 10.3389/fimmu.2018.03006] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
Malaria caused by the Plasmodium family of parasites, especially P.falciparum and P. vivax, is a major health problem in many countries in the tropical and subtropical regions of the world. The disease presents a wide array of systemic clinical conditions and several life-threatening organ pathologies, including the dreaded cerebral malaria. Like many other infectious diseases, malaria is an inflammatory response-driven disease, and positive outcomes to infection depend on finely tuned regulation of immune responses that efficiently clear parasites and allow protective immunity to develop. Immune responses initiated by the innate immune system in response to parasites play key roles both in protective immunity development and pathogenesis. Initial pro-inflammatory responses are essential for clearing infection by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged pro-inflammatory responses owing to inappropriate cellular programming contribute to disease conditions. A comprehensive knowledge of the molecular and cellular mechanisms that initiate immune responses and how these responses contribute to protective immunity development or pathogenesis is important for developing effective therapeutics and/or a vaccine. Historically, in efforts to develop a vaccine, immunity to malaria was extensively studied in the context of identifying protective humoral responses, targeting proteins involved in parasite invasion or clearance. The innate immune response was thought to be non-specific. However, during the past two decades, there has been a significant progress in understanding the molecular and cellular mechanisms of host-parasite interactions and the associated signaling in immune responses to malaria. Malaria infection occurs at two stages, initially in the liver through the bite of a mosquito, carrying sporozoites, and subsequently, in the blood through the invasion of red blood cells by merozoites released from the infected hepatocytes. Soon after infection, both the liver and blood stage parasites are sensed by various receptors of the host innate immune system resulting in the activation of signaling pathways and production of cytokines and chemokines. These immune responses play crucial roles in clearing parasites and regulating adaptive immunity. Here, we summarize the knowledge on molecular mechanisms that underlie the innate immune responses to malaria infection.
Collapse
Affiliation(s)
- D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
17
|
Zhang Y, Shao L. Decreased microRNA-140-5p contributes to respiratory syncytial virus disease through targeting Toll-like receptor 4. Exp Ther Med 2018; 16:993-999. [PMID: 30116350 DOI: 10.3892/etm.2018.6272] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 08/28/2017] [Indexed: 12/24/2022] Open
Abstract
The abnormal expression of miRNAs (miRs) has previously been reported in respiratory syncytial virus (RSV) disease. However, to the best of our knowledge, the expression of miR-140-5p in patients with an RSV infection has never been explored. Reverse transcription-quantitative polymerase chain reaction was performed to analyze the level of miR-140-5p in the blood and nasopharyngeal airway samples. ELISAs were performed to determine the levels of tumor necrosis factor α, interleukin (IL)-1β, IL-6 and IL-8. A dual luciferase reporter assay was also performed to investigate the possible target gene of miR-140-5p. The results demonstrated that the levels of miR-140-5p were significantly decreased in the nasal mucosal and peripheral blood samples of patients with RSV infection. It was also revealed that overexpression of miR-140-5p decreased the inflammatory responses, while inhibition of miR-140-5p enhanced the inflammatory responses. Additionally, three binding sites of miR-140-5p in the 3untranslated region (UTR) of Toll-like receptor (TLR)4 were identified and a dual luciferase reporter assay demonstrated that miR-140-5p significantly suppressed the relative luciferase activity of pmirGLO-TLR4-3UTR. Furthermore, the level of miR-140-5p was shown to be increased following interferon (IFN)α incubation. Notably, inhibition of miR-140-5p markedly attenuated IFNα-mediated downregulation of tumor necrosis factor α, and interleukin-1β, -6 and -8 in BEAS-2B cells. In summary, decreased miR-140-5p levels are involved in RSV-infection diseases primarily through targeting TLR4.
Collapse
Affiliation(s)
- Yun Zhang
- Infectious Disease Department, Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Lingyun Shao
- Infectious Disease Department, Shanghai Huashan Hospital, Shanghai 200040, P.R. China
| |
Collapse
|
18
|
DeFilippo J, Ebersole J, Beck G. Comparison of phagocytosis in three Caribbean Sea urchins. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 78:14-25. [PMID: 28916267 DOI: 10.1016/j.dci.2017.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/24/2017] [Accepted: 09/10/2017] [Indexed: 06/07/2023]
Abstract
In 1983 large numbers of the sea urchin Diadema antillarum unexplainably began showing signs of illness and dying in the Caribbean, and over the next year they came close to extinction, making it one of the worst mass mortality events on record. Present evidence suggests a water-borne pathogen as the etiological agent. Decades later Diadema densities remain low, and its near extinction has been a major factor in transforming living coral reefs in the Caribbean to barren algae-covered rock. In the ensuing decades, no solid explanation has been found to the questions: what killed Diadema; why did Diadema succumb while other species of urchins on the same reefs did not; and why has Diadema still not recovered? A recent hypothesis posited by our lab as to Diadema's vulnerability was directed at possible compromised immunity in Diadema, and experimental results found a significantly impaired humoral response to a key component of gram-negative bacteria. Here we use flow cytometry to examine the cellular arm of invertebrate immunity. We performed cytotoxicity and phagocytosis assays as a measure of the cellular immune responses of cells from Diadema and two other species of sea urchins not affected by the die-off. Despite our previous findings of in impaired humoral response, our study found no apparent difference in the cellular phagocytic response of Diadema compared to the other urchin species studied.
Collapse
Affiliation(s)
- John DeFilippo
- Department of Biology, University of Massachusetts at Boston, Boston, MA, 02125-3393, USA
| | - John Ebersole
- Department of Biology, University of Massachusetts at Boston, Boston, MA, 02125-3393, USA
| | - Gregory Beck
- Department of Biology, University of Massachusetts at Boston, Boston, MA, 02125-3393, USA.
| |
Collapse
|
19
|
Taghavi M, Khosravi A, Mortaz E, Nikaein D, Athari SS. Role of pathogen-associated molecular patterns (PAMPS) in immune responses to fungal infections. Eur J Pharmacol 2017; 808:8-13. [DOI: 10.1016/j.ejphar.2016.11.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 10/22/2016] [Accepted: 11/08/2016] [Indexed: 12/26/2022]
|
20
|
Levin R, Grinstein S, Canton J. The life cycle of phagosomes: formation, maturation, and resolution. Immunol Rev 2017; 273:156-79. [PMID: 27558334 DOI: 10.1111/imr.12439] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phagocytosis, the regulated uptake of large particles (>0.5 μm in diameter), is essential for tissue homeostasis and is also an early, critical component of the innate immune response. Phagocytosis can be conceptually divided into three stages: phagosome, formation, maturation, and resolution. Each of these involves multiple reactions that require exquisite spatial and temporal orchestration. The molecular events underlying these stages are being unraveled and the current state of knowledge is briefly summarized in this article.
Collapse
Affiliation(s)
- Roni Levin
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Johnathan Canton
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
21
|
Bi J, Koivisto L, Pang A, Li M, Jiang G, Aurora S, Wang Z, Owen GR, Dai J, Shen Y, Grenier D, Haapasalo M, Häkkinen L, Larjava H. Suppression of αvβ6 Integrin Expression by Polymicrobial Oral Biofilms in Gingival Epithelial Cells. Sci Rep 2017; 7:4411. [PMID: 28667248 PMCID: PMC5493688 DOI: 10.1038/s41598-017-03619-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/02/2017] [Indexed: 12/30/2022] Open
Abstract
Periodontal diseases manifest by the formation of deep pockets between the gingiva and teeth where multispecies bacterial biofilms flourish, causing inflammation and bone loss. Epithelial cell receptor αvβ6 integrin that regulates inflammation by activating the anti-inflammatory cytokine transforming growth factor-β1, is highly expressed in healthy junctional epithelium that connects the gingiva to the tooth enamel. However, its expression is attenuated in human periodontal disease. Moreover, Itgb6−/− mice display increased periodontal inflammation compared to wild-type mice. We hypothesized that bacterial biofilms present in the periodontal pockets suppress αvβ6 integrin levels in periodontal disease and that this change aggravates inflammation. To this end, we generated three-week-old multi-species oral biofilms in vitro and treated cultured gingival epithelial cells (GECs) with their extracts. The biofilm extracts caused suppression of β6 integrin expression and upregulation of pro-inflammatory cytokines, including interleukin-1β and -6. Furthermore, GECs with β6 integrin siRNA knockdown showed increased interleukin-1β expression, indicating that αvβ6 integrin-deficiency is associated with pro-inflammatory cytokine responsiveness. FSL-1, a synthetic bacterial lipopeptide, also suppressed β6 integrin expression in GECs. Therefore, biofilm components, including lipopeptides, may downregulate αvβ6 integrin expression in the pocket epithelium and thus promote epithelial cell-driven pro-inflammatory response in periodontal disease.
Collapse
Affiliation(s)
- Jiarui Bi
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Leeni Koivisto
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Aihui Pang
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming Li
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoqiao Jiang
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Saljae Aurora
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zhejun Wang
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Gethin R Owen
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jiayin Dai
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Ya Shen
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Daniel Grenier
- Faculté de Médecine Dentaire, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Markus Haapasalo
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lari Häkkinen
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hannu Larjava
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
22
|
Waldmannová E, Caisová V, Fáberová J, Sváčková P, Kovářová M, Sváčková D, Kumžáková Z, Jačková A, Vácová N, Nedbalová P, Horká M, Kopecký J, Ženka J. The use of Zymosan A and bacteria anchored to tumor cells for effective cancer immunotherapy: B16-F10 murine melanoma model. Int Immunopharmacol 2016; 39:295-306. [DOI: 10.1016/j.intimp.2016.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 12/27/2022]
|
23
|
Frikeche J, Maiti G, Chakravarti S. Small leucine-rich repeat proteoglycans in corneal inflammation and wound healing. Exp Eye Res 2016; 151:142-9. [PMID: 27569372 DOI: 10.1016/j.exer.2016.08.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
The small leucine rich repeat proteoglycans are major components of the cornea. Lumican, keratocan, decorin, biglycan and osteoglycin are present throughout the adult corneal stroma, and fibromodulin in the peripheral limbal area. In the cornea literature these proteoglycan have been reviewed as structural, collagen fibril-regulating proteins of the cornea. However, these proteoglycans are members of the leucine-rich-repeat superfamily, and share structural similarities with pathogen recognition toll-like receptors. Emerging studies are showing that these have a range of interactions with cell surface receptors, chemokines, growth factors and pathogen associated molecular patterns and are able to regulate host immune response, inflammation and wound healing. This review discusses what is known about their innate immune-related role directly in the cornea, and studies outside the field that find interesting links with innate immune and wound healing responses that are likely to be relevant to the ocular surface. In addition, the review discusses phenotypes of mice with targeted deletion of proteoglycan genes and genetic variants associated with human pathologies.
Collapse
Affiliation(s)
- Jihane Frikeche
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - George Maiti
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Shukti Chakravarti
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, USA; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, USA; Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, USA.
| |
Collapse
|
24
|
Sigola LB, Fuentes AL, Millis LM, Vapenik J, Murira A. Effects of Toll-like receptor ligands on RAW 264.7 macrophage morphology and zymosan phagocytosis. Tissue Cell 2016; 48:389-96. [PMID: 27157550 DOI: 10.1016/j.tice.2016.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 02/01/2016] [Accepted: 04/21/2016] [Indexed: 01/21/2023]
Abstract
In this study we compared the effects of the Toll-like receptor (TLR) ligands lipopolysaccharide (LPS), flagellin, the synthetic bacterial triacylated lipopeptide Pam3-Cys-Ser-Lys4 (Pam3CSK4), Polyinosinic:polycytidylic acid (Poly I:C), and macrophage-activating lipopeptide (MALP-2), which are TLR4, TLR5, TLR1/2, TLR3, and TLR2/6 agonists, respectively, on cell morphology and phagocytosis of zymosan particles, derived from Saccharomyces cerevisiae, and rich in fungal PAMPs including beta-glucan, mannose, and chitin. LPS, Pam3CSK4, and MALP-2 induced an activated macrophage phenotype and enhanced zymosan phagocytosis. In contrast, flagellin and Poly I:C, respectively, had little effect on cell morphology and phagocytosis. We examined the role of scavenger receptor A (SR-A) on zymosan phagocytosis. Cells cultured in medium alone expressed SR-A, and LPS induced further expression of the receptor. We also observed inhibitory effects of scavenger receptor antagonists fucoidan, dextran sulphate, and Polyinosinic (Poly I), respectively, on zymosan phagocytosis of cells in medium alone and those pre-treated with LPS. We conclude that exposure to specific TLR ligands impacts both cellular morphology and phagocytic capacity, and that scavenger receptors contribute to zymosan ingestion as well as LPS-induced augmentation of phagocytosis.
Collapse
Affiliation(s)
- Lynette B Sigola
- Biology Department, Douglas College, P.O. Box 2503, New Westminster B.C. V3L 5B2, Canada.
| | - Ana-Lucía Fuentes
- Biology Department, Douglas College, P.O. Box 2503, New Westminster B.C. V3L 5B2, Canada
| | - Leonard M Millis
- Biology Department, Douglas College, P.O. Box 2503, New Westminster B.C. V3L 5B2, Canada
| | - Jacqueline Vapenik
- Faculty of Health Sciences, Douglas College, P.O. Box 2503, New Westminster B.C. V3L 5B2, Canada
| | - Armstrong Murira
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, B.C. V5A 1S6 Canada
| |
Collapse
|
25
|
Billings EA, Lee CS, Owen KA, D'Souza RS, Ravichandran KS, Casanova JE. The adhesion GPCR BAI1 mediates macrophage ROS production and microbicidal activity against Gram-negative bacteria. Sci Signal 2016; 9:ra14. [PMID: 26838550 PMCID: PMC4894535 DOI: 10.1126/scisignal.aac6250] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The detection of microbes and initiation of an innate immune response occur through pattern recognition receptors (PRRs), which are critical for the production of inflammatory cytokines and activation of the cellular microbicidal machinery. In particular, the production of reactive oxygen species (ROS) by the NADPH oxidase complex is a critical component of the macrophage bactericidal machinery. We previously characterized brain-specific angiogenesis inhibitor 1 (BAI1), a member of the adhesion family of G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs), as a PRR that mediates the selective phagocytic uptake of Gram-negative bacteria by macrophages. We showed that BAI1 promoted phagosomal ROS production through activation of the Rho family guanosine triphosphatase (GTPase) Rac1, thereby stimulating NADPH oxidase activity. Primary BAI1-deficient macrophages exhibited attenuated Rac GTPase activity and reduced ROS production in response to several Gram-negative bacteria, resulting in impaired microbicidal activity. Furthermore, in a peritoneal infection model, BAI1-deficient mice exhibited increased susceptibility to death by bacterial challenge because of impaired bacterial clearance. Together, these findings suggest that BAI1 mediates the clearance of Gram-negative bacteria by stimulating both phagocytosis and NADPH oxidase activation, thereby coupling bacterial detection to the cellular microbicidal machinery.
Collapse
Affiliation(s)
- Emily A Billings
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Chang Sup Lee
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Katherine A Owen
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ryan S D'Souza
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - James E Casanova
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA. Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
26
|
Shi M, Zhang Y, Liu L, Zhang T, Han F, Cleveland J, Wang F, McKeehan WL, Li Y, Zhang D. MAP1S Protein Regulates the Phagocytosis of Bacteria and Toll-like Receptor (TLR) Signaling. J Biol Chem 2016; 291:1243-50. [PMID: 26565030 PMCID: PMC4714212 DOI: 10.1074/jbc.m115.687376] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/02/2015] [Indexed: 12/29/2022] Open
Abstract
Phagocytosis is a critical cellular process for innate immune defense against microbial infection. The regulation of phagocytosis process is complex and has not been well defined. An intracellular molecule might regulate cell surface-initiated phagocytosis, but the underlying molecular mechanism is poorly understood (1). In this study, we found that microtubule-associated protein 1S (MAP1S), a protein identified recently that is involved in autophagy (2), is expressed primarily in macrophages. MAP1S-deficient macrophages are impaired in the phagocytosis of bacteria. Furthermore, we demonstrate that MAP1S interacts directly with MyD88, a key adaptor of Toll-like receptors (TLRs), upon TLR activation and affects the TLR signaling pathway. Intriguingly, we also observe that, upon TLR activation, MyD88 participates in autophagy processing in a MAP1S-dependent manner by co-localizing with MAP1 light chain 3 (MAP1-LC3 or LC3). Therefore, we reveal that an intracellular autophagy-related molecule of MAP1S controls bacterial phagocytosis through TLR signaling.
Collapse
Affiliation(s)
- Ming Shi
- From the Center for Infectious and Inflammatory Diseases and Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030 and
| | - Yifan Zhang
- From the Center for Infectious and Inflammatory Diseases and
| | - Leyuan Liu
- the School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Tingting Zhang
- From the Center for Infectious and Inflammatory Diseases and
| | - Fang Han
- From the Center for Infectious and Inflammatory Diseases and Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030 and
| | | | - Fen Wang
- the School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Wallace L McKeehan
- the School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yu Li
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, Texas 77030 and
| | - Dekai Zhang
- From the Center for Infectious and Inflammatory Diseases and
| |
Collapse
|
27
|
Staphylococcus aureus Biofilms Induce Macrophage Dysfunction Through Leukocidin AB and Alpha-Toxin. mBio 2015; 6:mBio.01021-15. [PMID: 26307164 PMCID: PMC4550693 DOI: 10.1128/mbio.01021-15] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The macrophage response to planktonic Staphylococcus aureus involves the induction of proinflammatory microbicidal activity. However, S. aureus biofilms can interfere with these responses in part by polarizing macrophages toward an anti-inflammatory profibrotic phenotype. Here we demonstrate that conditioned medium from mature S. aureus biofilms inhibited macrophage phagocytosis and induced cytotoxicity, suggesting the involvement of a secreted factor(s). Iterative testing found the active factor(s) to be proteinaceous and partially agr-dependent. Quantitative mass spectrometry identified alpha-toxin (Hla) and leukocidin AB (LukAB) as critical molecules secreted by S. aureus biofilms that inhibit murine macrophage phagocytosis and promote cytotoxicity. A role for Hla and LukAB was confirmed by using hla and lukAB mutants, and synergy between the two toxins was demonstrated with a lukAB hla double mutant and verified by complementation. Independent confirmation of the effects of Hla and LukAB on macrophage dysfunction was demonstrated by using an isogenic strain in which Hla was constitutively expressed, an Hla antibody to block toxin activity, and purified LukAB peptide. The importance of Hla and LukAB during S. aureus biofilm formation in vivo was assessed by using a murine orthopedic implant biofilm infection model in which the lukAB hla double mutant displayed significantly lower bacterial burdens and more macrophage infiltrates than each single mutant. Collectively, these findings reveal a critical synergistic role for Hla and LukAB in promoting macrophage dysfunction and facilitating S. aureus biofilm development in vivo. Staphylococcus aureus has a propensity to form multicellular communities known as biofilms. While growing in a biofilm, S. aureus displays increased tolerance to nutrient deprivation, antibiotic insult, and even host immune challenge. Previous studies have shown that S. aureus biofilms thwart host immunity in part by preventing macrophage phagocytosis. It remained unclear whether this was influenced solely by the considerable size of biofilms or whether molecules were also actively secreted to circumvent macrophage-mediated phagocytosis. This is the first report to demonstrate that S. aureus biofilms inhibit macrophage phagocytosis and induce macrophage death through the combined action of leukocidin AB and alpha-toxin. Loss of leukocidin AB and alpha-toxin expression resulted in enhanced S. aureus biofilm clearance in a mouse model of orthopedic implant infection, suggesting that these toxins could be targeted therapeutically to facilitate biofilm clearance in humans.
Collapse
|
28
|
Scherr TD, Heim CE, Morrison JM, Kielian T. Hiding in Plain Sight: Interplay between Staphylococcal Biofilms and Host Immunity. Front Immunol 2014; 5:37. [PMID: 24550921 PMCID: PMC3913997 DOI: 10.3389/fimmu.2014.00037] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/22/2014] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are notable for their propensity to form biofilms on implanted medical devices. Staphylococcal biofilm infections are typified by their recalcitrance to antibiotics and ability to circumvent host immune-mediated clearance, resulting in the establishment of chronic infections that are often recurrent in nature. Indeed, the immunomodulatory lifestyle of biofilms seemingly shapes the host immune response to ensure biofilm engraftment and persistence in an immune competent host. Here, we provide a brief review of the mechanisms whereby S. aureus and S. epidermidis biofilms manipulate host–pathogen interactions and discuss the concept of microenvironment maintenance in infectious outcomes, as well as speculate how these findings pertain to the challenges of staphylococcal vaccine development.
Collapse
Affiliation(s)
- Tyler D Scherr
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - John M Morrison
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| |
Collapse
|
29
|
Janotová T, Jalovecká M, Auerová M, Švecová I, Bruzlová P, Maierová V, Kumžáková Z, Čunátová Š, Vlčková Z, Caisová V, Rozsypalová P, Lukáčová K, Vácová N, Wachtlová M, Salát J, Lieskovská J, Kopecký J, Ženka J. The use of anchored agonists of phagocytic receptors for cancer immunotherapy: B16-F10 murine melanoma model. PLoS One 2014; 9:e85222. [PMID: 24454822 PMCID: PMC3890306 DOI: 10.1371/journal.pone.0085222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/25/2013] [Indexed: 01/03/2023] Open
Abstract
The application of the phagocytic receptor agonists in cancer immunotherapy was studied. Agonists (laminarin, molecules with terminal mannose, N-Formyl-methioninyl-leucyl-phenylalanine) were firmly anchored to the tumor cell surface. When particular agonists of phagocytic receptors were used together with LPS (Toll-like receptor agonist), high synergy causing tumour shrinkage and a temporary or permanent disappearance was observed. Methods of anchoring phagocytic receptor agonists (charge interactions, anchoring based on hydrophobic chains, covalent bonds) and various regimes of phagocytic agonist/LPS mixture applications were tested to achieve maximum therapeutic effect. Combinations of mannan/LPS and f-MLF/LPS (hydrophobic anchors) in appropriate (pulse) regimes resulted in an 80% and 60% recovery for mice, respectively. We propose that substantial synergy between agonists of phagocytic and Toll-like receptors (TLR) is based on two events. The TLR ligand induces early and massive inflammatory infiltration of tumors. The effect of this cell infiltrate is directed towards tumor cells, bearing agonists of phagocytic receptors on their surface. The result of these processes was effective killing of tumor cells. This novel approach represents exploitation of innate immunity mechanisms for treating cancer.
Collapse
Affiliation(s)
- Tereza Janotová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Marie Jalovecká
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Marie Auerová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Ivana Švecová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Pavlína Bruzlová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Veronika Maierová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Zuzana Kumžáková
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Štěpánka Čunátová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Zuzana Vlčková
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Veronika Caisová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Petra Rozsypalová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Katarína Lukáčová
- Department of Pathology, Regional Hospital, České Budějovice, Czech Republic
| | - Nikol Vácová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Markéta Wachtlová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Jiří Salát
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic
| | - Jaroslava Lieskovská
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Jan Kopecký
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Jan Ženka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
- * E-mail:
| |
Collapse
|
30
|
Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Prog Neurobiol 2013; 115:25-44. [PMID: 24291544 DOI: 10.1016/j.pneurobio.2013.11.003] [Citation(s) in RCA: 454] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 02/07/2023]
Abstract
Intracerebral hemorrhage (ICH) accounts for 10-15% of all strokes and is associated with high mortality and morbidity. Currently, no effective medical treatment is available to improve functional outcomes in patients with ICH. Potential therapies targeting secondary brain injury are arousing a great deal of interest in translational studies. Increasing evidence has shown that inflammation is the key contributor of ICH-induced secondary brain injury. Inflammation progresses in response to various stimuli produced after ICH. Hematoma components initiate inflammatory signaling via activation of microglia, subsequently releasing proinflammatory cytokines and chemokines to attract peripheral inflammatory infiltration. Hemoglobin (Hb), heme, and iron released after red blood cell lysis aggravate ICH-induced inflammatory injury. Danger associated molecular patterns such as high mobility group box 1 protein, released from damaged or dead cells, trigger inflammation in the late stage of ICH. Preclinical studies have identified inflammatory signaling pathways that are involved in microglial activation, leukocyte infiltration, toll-like receptor (TLR) activation, and danger associated molecular pattern regulation in ICH. Recent advances in understanding the pathogenesis of ICH-induced inflammatory injury have facilitated the identification of several novel therapeutic targets for the treatment of ICH. This review summarizes recent progress concerning the mechanisms underlying ICH-induced inflammation. We focus on the inflammatory signaling pathways involved in microglial activation and TLR signaling, and explore potential therapeutic interventions by targeting the removal of hematoma components and inhibition of TLR signaling.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yanchun Wang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
31
|
Lima WC, Balestrino D, Forestier C, Cosson P. Two distinct sensing pathways allow recognition of Klebsiella pneumoniae by Dictyostelium amoebae. Cell Microbiol 2013; 16:311-23. [PMID: 24128258 PMCID: PMC4303882 DOI: 10.1111/cmi.12226] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/24/2013] [Accepted: 10/07/2013] [Indexed: 12/24/2022]
Abstract
Recognition of bacteria by metazoans is mediated by receptors that recognize different types of microorganisms and elicit specific cellular responses. The soil amoebae Dictyostelium discoideum feeds upon a variable mixture of environmental bacteria, and it is expected to recognize and adapt to various food sources. To date, however, no bacteria-sensing mechanisms have been described. In this study, we isolated a Dictyostelium mutant (fspA KO) unable to grow in the presence of non-capsulated Klebsiella pneumoniae bacteria, but growing as efficiently as wild-type cells in the presence of other bacteria, such as Bacillus subtilis. fspA KO cells were also unable to respond to K. pneumoniae and more specifically to bacterially secreted folate in a chemokinetic assay, while they responded readily to B. subtilis. Remarkably, both WT and fspA KO cells were able to grow in the presence of capsulated LM21 K. pneumoniae, and responded to purified capsule, indicating that capsule recognition may represent an alternative, FspA-independent mechanism for K. pneumoniae sensing. When LM21 capsule synthesis genes were deleted, growth and chemokinetic response were lost for fspA KO cells, but not for WT cells. Altogether, these results indicate that Dictyostelium amoebae use specific recognition mechanisms to respond to different K. pneumoniae elements.
Collapse
Affiliation(s)
- Wanessa C Lima
- Department for Cell Physiology and Metabolism, Centre Medical Universitaire, University of Geneva, 1 rue Michel Servet, 1211, Geneva 4, Switzerland
| | | | | | | |
Collapse
|
32
|
van Bergenhenegouwen J, Plantinga TS, Joosten LAB, Netea MG, Folkerts G, Kraneveld AD, Garssen J, Vos AP. TLR2 & Co: a critical analysis of the complex interactions between TLR2 and coreceptors. J Leukoc Biol 2013; 94:885-902. [PMID: 23990624 DOI: 10.1189/jlb.0113003] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
TLRs play a major role in microbe-host interactions and innate immunity. Of the 10 functional TLRs described in humans, TLR2 is unique in its requirement to form heterodimers with TLR1 or TLR6 for the initiation of signaling and cellular activation. The ligand specificity of TLR2 heterodimers has been studied extensively, using specific bacterial and synthetic lipoproteins to gain insight into the structure-function relationship, the minimal active motifs, and the critical dependence on TLR1 or TLR6 for activation. Different from that for specific well-defined TLR2 agonists, recognition of more complex ligands like intact microbes or molecules from endogenous origin requires TLR2 to interact with additional coreceptors. A breadth of data has been published on ligand-induced interactions of TLR2 with additional pattern recognition receptors such as CD14, scavenger receptors, integrins, and a range of other receptors, all of them important factors in TLR2 function. This review summarizes the roles of TLR2 in vivo and in specific immune cell types and integrates this information with a detailed review of our current understanding of the roles of specific coreceptors and ligands in regulating TLR2 functions. Understanding how these processes affect intracellular signaling and drive functional immune responses will lead to a better understanding of host-microbe interactions and will aid in the design of new agents to target TLR2 function in health and disease.
Collapse
|
33
|
Li J, Fang L, Killer HE, Flammer J, Meyer P, Neutzner A. Meningothelial cells as part of the central nervous system host defence. Biol Cell 2013; 105:304-15. [PMID: 23634770 DOI: 10.1111/boc.201300013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 04/19/2013] [Indexed: 01/29/2023]
Abstract
BACKGROUND INFORMATION Meningothelial cells (MECs) are the cellular components of the meninges protecting the brain and as such provide important barrier function for the central nervous system building the interface between neuronal tissue and the cerebrospinal fluid (CSF). MECs were previously shown to be involved in the clearance of waste products from the CSF and in maintaining the optic nerve microenvironment. In addition, MECs are involved in immunological processes in the brain by secretion of pro-inflammatory cytokines in response to various pathologically relevant stress conditions. RESULTS In this study, we analysed the uptake of latex beads as well as bacteria by human MECs using flow cytometric analyses. We found that MECs are highly active phagocytes able of ingesting large amounts of latex beads, as well as Gram-positive and Gram-negative bacteria. Phagocytic activity of MECs was sensitive to nocodazole and cytochalasin D treatment to a varying degree depending on particle composition. Interestingly, Gram-positive bacteria such as Staphylococcus aureus are more readily taken up compared with Gram-negative Escherichia coli. In addition, pre-treatment of MECs with lipopolysaccharide (LPS) or phorbol-12-myristate-13-acetate (PMA) enhanced S. aureus uptake, whereas PMA but not LPS was effective in enhancing E. coli uptake. CONCLUSIONS Thus, MECs are highly active facultative phagocytes likely important for the maintenance of CSF homeostasis and host defence in the central nervous system especially against Gram-positive bacteria.
Collapse
Affiliation(s)
- Jia Li
- Department of Biomedicine, Ocular Pharmacology and Physiology, University Basel, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
34
|
Sokolovska A, Becker CE, Ip WKE, Rathinam VAK, Brudner M, Paquette N, Tanne A, Vanaja SK, Moore KJ, Fitzgerald KA, Lacy-Hulbert A, Stuart LM. Activation of caspase-1 by the NLRP3 inflammasome regulates the NADPH oxidase NOX2 to control phagosome function. Nat Immunol 2013; 14:543-53. [PMID: 23644505 PMCID: PMC3708594 DOI: 10.1038/ni.2595] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/28/2013] [Indexed: 11/10/2022]
Abstract
Phagocytosis is a fundamental cellular process that is pivotal for immunity as it coordinates microbial killing, innate immune activation and antigen presentation. An essential step in this process is phagosome acidification, which regulates many functions of these organelles that allow phagosomes to participate in processes that are essential to both innate and adaptive immunity. Here we report that acidification of phagosomes containing Gram-positive bacteria is regulated by the NLRP3 inflammasome and caspase-1. Active caspase-1 accumulates on phagosomes and acts locally to control the pH by modulating buffering by the NADPH oxidase NOX2. These data provide insight into a mechanism by which innate immune signals can modify cellular defenses and establish a new function for the NLRP3 inflammasome and caspase-1 in host defense.
Collapse
Affiliation(s)
- Anna Sokolovska
- Developmental Immunology and Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pál L, Árnyas EM, Tóth B, Ádám B, Rácz G, Ádány R, McKee M, Szűcs S. Aliphatic alcohol contaminants of illegally produced spirits inhibit phagocytosis by human granulocytes. Immunopharmacol Immunotoxicol 2013; 35:251-6. [DOI: 10.3109/08923973.2012.759962] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
36
|
Toll-like receptor 4 in phagocytosis of Escherichia coli by endotoxin-activated human neutrophils in whole blood. Crit Care 2012. [PMCID: PMC3504881 DOI: 10.1186/cc11767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
37
|
Listeria monocytogenes-derived listeriolysin O has pathogen-associated molecular pattern-like properties independent of its hemolytic ability. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 20:77-84. [PMID: 23136118 DOI: 10.1128/cvi.00488-12] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
There is a constant need for improved adjuvants to augment the induction of immune responses against tumor-associated antigens (TAA) during immunotherapy. Previous studies have established that listeriolysin O (LLO), a cholesterol-dependent cytolysin derived from Listeria monocytogenes, exhibits multifaceted effects to boost the stimulation of immune responses to a variety of antigens. However, the direct ability of LLO as an adjuvant and whether it acts as a pathogen-associated molecular pattern (PAMP) have not been demonstrated. In this paper, we show that a detoxified, nonhemolytic form of LLO (dtLLO) is an effective adjuvant in tumor immunotherapy and may activate innate and cellular immune responses by acting as a PAMP. Our investigation of the adjuvant activity demonstrates that dtLLO, either fused to or administered as a mixture with a human papillomavirus type 16 (HPV-16) E7 recombinant protein, can augment antitumor immune responses and facilitate tumor eradication. Further mechanistic studies using bone marrow-derived dendritic cells suggest that dtLLO acts as a PAMP by stimulating production of proinflammatory cytokines and inducing maturation of antigen-presenting cells (APC). We propose that dtLLO is an effective adjuvant for tumor immunotherapy, and likely for other therapeutic settings.
Collapse
|
38
|
Kumar S, Gowda NM, Wu X, Gowda RN, Gowda DC. CD36 modulates proinflammatory cytokine responses to Plasmodium falciparum glycosylphosphatidylinositols and merozoites by dendritic cells. Parasite Immunol 2012; 34:372-82. [PMID: 22486596 DOI: 10.1111/j.1365-3024.2012.01367.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies have shown that glycosylphosphatidylinositols (GPIs) of Plasmodium falciparum activate macrophages mainly through Toll-like receptor 2 (TLR2)-mediated signalling and to certain extent through TLR4-mediated signalling to induce proinflammatory cytokine production. However, the ability of parasite GPIs to activate dendritic cells (DCs) has not been reported. Here, we show that parasite GPIs efficiently activate DCs through TLR2-mediated signalling mechanism and induce the production of TNF-α and IL-12. We also studied the role of scavenger receptor CD36 in P. falciparum GPI- and merozoite-induced cytokine responses by DCs. The results indicate that CD36 modulates the cytokine-inducing activity of the parasite GPIs by collaborating with TLR2 in DCs. Furthermore, our data reveal that CD36 modulates the activity of P. falciparum merozoites, likely by the contribution of phagocytosis-coupled CD36-mediated signalling to the signalling induced by merozoites. Altogether, these results contribute towards understanding of signalling mechanisms in malaria parasite-induced activation of the innate immune system.
Collapse
Affiliation(s)
- S Kumar
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
39
|
Shao H, Lee S, Gae-Scott S, Nakata C, Chen S, Hamad AR, Chakravarti S. Extracellular matrix lumican promotes bacterial phagocytosis, and Lum-/- mice show increased Pseudomonas aeruginosa lung infection severity. J Biol Chem 2012; 287:35860-72. [PMID: 22865855 DOI: 10.1074/jbc.m112.380550] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Phagocytosis is central to bacterial clearance, but the exact mechanism is incompletely understood. Here, we show a novel and critical role for lumican, the connective tissue extracellular matrix small leucine-rich repeat proteoglycan, in CD14-mediated bacterial phagocytosis. In Psuedomonas aeruginosa lung infections, lumican-deficient (Lum(-/-)) mice failed to clear the bacterium from lungs, tissues, and showed a dramatic increase in mortality. In vitro, phagocytosis of nonopsonized gram-negative Escherichia coli and P. aeruginosa was inhibited in Lum(-/-) peritoneal macrophages (MΦs). Lumican co-localized with CD14, CD18, and bacteria on Lum(+/+) MΦ surfaces. Using two different P. aeruginosa strains that require host CD14 (808) or CD18/CR3 (P1) for phagocytosis, we showed that lumican has a larger role in CD14-mediated phagocytosis. Recombinant lumican (rLum) restored phagocytosis in Lum(-/-) MΦs. Surface plasmon resonance showed specific binding of rLum to CD14 (K(A) = 2.15 × 10(6) M(-1)), whereas rLumY20A, and not rLumY21A, where a tyrosine in each was replaced with an alanine, showed 60-fold decreased binding. The rLumY20A variant also failed to restore phagocytosis in Lum(-/-) MΦs, indicating Tyr-20 to be functionally important. Thus, in addition to a structural role in connective tissues, lumican has a major protective role in gram-negative bacterial infections, a novel function for small leucine-rich repeat proteoglycans.
Collapse
Affiliation(s)
- HanJuan Shao
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Guzman-Beltran S, Perez-Torres A, Coronel-Cruz C, Torres-Guerrero H. Phagocytic receptors on macrophages distinguish between different Sporothrix schenckii morphotypes. Microbes Infect 2012; 14:1093-101. [PMID: 22771955 DOI: 10.1016/j.micinf.2012.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/21/2012] [Accepted: 06/05/2012] [Indexed: 12/20/2022]
Abstract
Sporothrix schenckii is a human pathogen that causes sporotrichosis, a cutaneous subacute or chronic mycosis. Little is known about the innate immune response and the receptors involved in host recognition and phagocytosis of S. schenckii. Here, we demonstrate that optimal phagocytosis of conidia and yeast is dependent on preimmune human serum opsonisation. THP-1 macrophages efficiently ingested opsonised conidia. Competition with D-mannose, methyl α-D-mannopyranoside, D-fucose, and N-acetyl glucosamine blocked this process, suggesting the involvement of the mannose receptor in binding and phagocytosis of opsonised conidia. Release of TNF-α was not stimulated by opsonised or non-opsonised conidia, although reactive oxygen species (ROS) were produced, resulting in the killing of conidia by THP-1 macrophages. Heat inactivation of the serum did not affect conidia internalization, which was markedly decreased for yeast cells, suggesting the role of complement components in yeast uptake. Conversely, release of TNF-α and production of ROS were induced by opsonised and non-opsonised yeast. These data demonstrate that THP-1 macrophages respond to opsonised conidia and yeast through different phagocytic receptors, inducing a differential cellular response. Conidia induces a poor pro-inflammatory response and lower rate of ROS-induced cell death, thereby enhancing the pathogen's survival.
Collapse
Affiliation(s)
- Silvia Guzman-Beltran
- Departamento de Microbiología en Investigación, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Calzada de Tlalpan 4502, Col. Sección XVI, México DF, Mexico
| | | | | | | |
Collapse
|
41
|
Pei G, Bronietzki M, Gutierrez MG. Immune regulation of Rab proteins expression and intracellular transport. J Leukoc Biol 2012; 92:41-50. [DOI: 10.1189/jlb.0212076] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
42
|
Chen YJ, Hsieh MY, Chang MY, Chen HC, Jan MS, Maa MC, Leu TH. Eps8 protein facilitates phagocytosis by increasing TLR4-MyD88 protein interaction in lipopolysaccharide-stimulated macrophages. J Biol Chem 2012; 287:18806-19. [PMID: 22493489 DOI: 10.1074/jbc.m112.340935] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Toll-like receptors (TLRs) are crucial in macrophage phagocytosis, which is pivotal in host innate immune response. However, the detailed mechanism is not fully defined. Here, we demonstrated that the induction of Src and Eps8 in LPS-treated macrophages was TLR4- and MyD88-dependent, and their attenuation reduced LPS-promoted phagocytosis. Confocal microscopy indicated the colocalization of Eps8 and TLR4 in the cytosol and at the phagosome. Consistently, both Eps8 and TLR4 were present in the same immunocomplex regardless of LPS stimulation. Inhibition of this complex formation by eps8 siRNA or overexpression of pleckstrin homology domain-truncated Eps8 (i.e. 261-p97(Eps8)) decreased LPS-induced TLR4-MyD88 interaction and the following activation of Src, focal adhesion kinase, and p38 MAPK. Importantly, attenuation of Eps8 impaired the bacterium-killing ability of macrophages. Thus, Eps8 is a key regulator of the LPS-stimulated TLR4-MyD88 interaction and contributes to macrophage phagocytosis.
Collapse
Affiliation(s)
- Yen-Jen Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | | | | | | | | | | | | |
Collapse
|
43
|
Peroxisome Proliferator-Activator Receptor γ: A Link between Macrophage CD36 and Inflammation in Malaria Infection. PPAR Res 2012; 2012:640769. [PMID: 22287954 PMCID: PMC3263638 DOI: 10.1155/2012/640769] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/19/2011] [Indexed: 12/21/2022] Open
Abstract
Severe malaria infection caused by Plasmodium falciparum is a global life-threatening disease and a leading cause of death worldwide. Intensive investigations have demonstrated that macrophages play crucial roles in control of inflammatory and immune responses and clearance of Plasmodium-falciparum-parasitized erythrocytes (PE). This paper focuses on how macrophage CD36 recognizes and internalizes PE and participates the inflammatory signaling in response to Plasmodium falciparum. In addition, recent advances in our current understanding of the biological actions of PPARγ on CD36 and malaria clearance from the hosts are highlighted.
Collapse
|
44
|
Buschow SI, Lasonder E, Szklarczyk R, Oud MM, de Vries IJM, Figdor CG. Unraveling the human dendritic cell phagosome proteome by organellar enrichment ranking. J Proteomics 2011; 75:1547-62. [PMID: 22146474 DOI: 10.1016/j.jprot.2011.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 11/18/2011] [Accepted: 11/22/2011] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DC) take up pathogens through phagocytosis and process them into protein and lipid fragments for presentation to T cells. So far, the proteome of the human DC phagosome, a detrimental compartment for antigen processing and presentation as well as for DC activation, remains largely uncharacterized. Here we have analyzed the protein composition of phagosomes from human monocyte-derived DC. For LC-MS/MS analysis we purified phagosomes from DC using latex beads targeted to DC-SIGN, and quantified proteins using a label-free method. We used organellar enrichment ranking (OER) to select proteins with a high potential to be relevant for phagosome function. The method compares phagosome protein abundance with protein abundance in whole DC. Phagosome enrichment indicates specific recruitment to the phagosome rather than co-purification or passive incorporation. Using OER we extracted the most enriched proteins that we further complemented with functionally associated proteins to define a set of 90 phagosomal proteins that included many proteins with established relevance on DC phagosomes as well as high potential novel candidates. We already experimentally confirmed phagosomal recruitment of Galectin-9, which has not been previously associated with phagocytosis, to both bead and pathogen containing phagosomes, suggesting a role for Galectin-9 in DC phagocytosis.
Collapse
Affiliation(s)
- Sonja I Buschow
- Department of Tumor Immunology at the Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Postbox 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Rigby KM, DeLeo FR. Neutrophils in innate host defense against Staphylococcus aureus infections. Semin Immunopathol 2011; 34:237-59. [PMID: 22080185 PMCID: PMC3271231 DOI: 10.1007/s00281-011-0295-3] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 10/14/2011] [Indexed: 12/29/2022]
Abstract
Staphylococcus aureus has been an important human pathogen throughout history and is currently a leading cause of bacterial infections worldwide. S. aureus has the unique ability to cause a continuum of diseases, ranging from minor skin infections to fatal necrotizing pneumonia. Moreover, the emergence of highly virulent, drug-resistant strains such as methicillin-resistant S. aureus in both healthcare and community settings is a major therapeutic concern. Neutrophils are the most prominent cellular component of the innate immune system and provide an essential primary defense against bacterial pathogens such as S. aureus. Neutrophils are rapidly recruited to sites of infection where they bind and ingest invading S. aureus, and this process triggers potent oxidative and non-oxidative antimicrobial killing mechanisms that serve to limit pathogen survival and dissemination. S. aureus has evolved numerous mechanisms to evade host defense strategies employed by neutrophils, including the ability to modulate normal neutrophil turnover, a process critical to the resolution of acute inflammation. Here we provide an overview of the role of neutrophils in host defense against bacterial pathogens and discuss strategies employed by S. aureus to circumvent neutrophil function.
Collapse
Affiliation(s)
- Kevin M Rigby
- Laboratory of Human Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA
| | | |
Collapse
|
46
|
Hanke ML, Kielian T. Toll-like receptors in health and disease in the brain: mechanisms and therapeutic potential. Clin Sci (Lond) 2011; 121:367-87. [PMID: 21745188 PMCID: PMC4231819 DOI: 10.1042/cs20110164] [Citation(s) in RCA: 375] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The discovery of mammalian TLRs (Toll-like receptors), first identified in 1997 based on their homology with Drosophila Toll, greatly altered our understanding of how the innate immune system recognizes and responds to diverse microbial pathogens. TLRs are evolutionarily conserved type I transmembrane proteins expressed in both immune and non-immune cells, and are typified by N-terminal leucine-rich repeats and a highly conserved C-terminal domain termed the TIR [Toll/interleukin (IL)-1 receptor] domain. Upon stimulation with their cognate ligands, TLR signalling elicits the production of cytokines, enzymes and other inflammatory mediators that can have an impact on several aspects of CNS (central nervous system) homoeostasis and pathology. For example, TLR signalling plays a crucial role in initiating host defence responses during CNS microbial infection. Furthermore, TLRs are targets for many adjuvants which help shape pathogen-specific adaptive immune responses in addition to triggering innate immunity. Our knowledge of TLR expression and function in the CNS has greatly expanded over the last decade, with new data revealing that TLRs also have an impact on non-infectious CNS diseases/injury. In particular, TLRs recognize a number of endogenous molecules liberated from damaged tissues and, as such, influence inflammatory responses during tissue injury and autoimmunity. In addition, recent studies have implicated TLR involvement during neurogenesis, and learning and memory in the absence of any underlying infectious aetiology. Owing to their presence and immune-regulatory role within the brain, TLRs represent an attractive therapeutic target for numerous CNS disorders and infectious diseases. However, it is clear that TLRs can exert either beneficial or detrimental effects in the CNS, which probably depend on the context of tissue homoeostasis or pathology. Therefore any potential therapeutic manipulation of TLRs will require an understanding of the signals governing specific CNS disorders to achieve tailored therapy.
Collapse
Affiliation(s)
- Mark L. Hanke
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
47
|
Fuentes AL, Millis L, Sigola LB. Laminarin, a soluble beta-glucan, inhibits macrophage phagocytosis of zymosan but has no effect on lipopolysaccharide mediated augmentation of phagocytosis. Int Immunopharmacol 2011; 11:1939-45. [PMID: 21856445 DOI: 10.1016/j.intimp.2011.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 08/02/2011] [Accepted: 08/03/2011] [Indexed: 11/18/2022]
Abstract
Phagocytosis is a fundamental aspect of innate resistance against microbes, including fungi. In this study we investigated the significance of beta-glucan on the surfaces of zymosan particles, derived from Saccharomyces cerevisiae, during phagocytosis by RAW 264.7 macrophages. Phagocytosis was assessed in vitro by macrophage exposure to zymosan particles followed by cell staining and light microscopy. Macrophage ingestion of zymosan was dependent on cellular recognition of the particles' beta-glucans since laminarin, a soluble beta-glucan, inhibited phagocytosis in a concentration dependent manner when added to cell cultures. In contrast, the presence of another carbohydrate, mannan, had no effect on zymosan phagocytosis by cells. In addition we showed that LPS and dexamethasone had opposing effects on phagocytosis of zymosan. LPS significantly augmented ingestion while in contrast dexamethasone, like laminarin, suppressed it. The LPS-enhanced ingestion of zymosan was insensitive to the presence of laminarin in cell cultures, however dexamethasone partially ameliorated the effects of LPS on phagocytosis. Our findings confirm beta-glucan as an important ligand identified by macrophages and required for zymosan phagocytosis in naïve cells, but not in cells previously exposed to LPS.
Collapse
Affiliation(s)
- Ana-Lucia Fuentes
- Biology Department, Douglas College, P.O. Box 2503, New Westminster, BC, Canada V3L 5B2
| | | | | |
Collapse
|
48
|
Anand PK. Exosomal membrane molecules are potent immune response modulators. Commun Integr Biol 2011; 3:405-8. [PMID: 21057626 DOI: 10.4161/cib.3.5.12474] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 05/25/2010] [Indexed: 12/12/2022] Open
Abstract
Exosomes are endosome-derived vesicles (40-100 nm) formed during the formation of multi-vesicular bodies (MVBs). Occasionally, the MVBs fuse with the plasma membrane releasing their intra-luminal vesicles into the extracellular media, which are then known as exosomes. Different cell types such as B-cells, dendritic cells, platelets, reticulocytes and macrophages can release exosomes and current research in this area is more focused towards exosomes released by antigen-presenting cells. Exosomes have recently been shown to be immunomodulatory and the mechanism of immune response initiation by them is beginning to emerge. Besides molecules present inside the lumen of exosomes, it has been suggested that certain exosomal membrane molecules can interact with their surface receptors on the target cells thereby inducing an immunomodulatory response. In this review, Hsp70 and galectin-5, two immunogenic molecules present on exosomal membrane, are discussed in detail for initiating this response.
Collapse
Affiliation(s)
- Paras K Anand
- Cell Biology and Biophysics Unit; European Molecular Biology Laboratory; Heidelberg, Germany
| |
Collapse
|
49
|
Kang HJ, Ha JM, Kim HS, Lee H, Kurokawa K, Lee BL. The role of phagocytosis in IL-8 production by human monocytes in response to lipoproteins on Staphylococcus aureus. Biochem Biophys Res Commun 2011; 406:449-53. [DOI: 10.1016/j.bbrc.2011.02.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 02/12/2011] [Indexed: 01/02/2023]
|
50
|
Hafner AM, Corthésy B, Textor M, Merkle HP. Tuning the immune response of dendritic cells to surface-assembled poly(I:C) on microspheres through synergistic interactions between phagocytic and TLR3 signaling. Biomaterials 2011; 32:2651-61. [PMID: 21216459 DOI: 10.1016/j.biomaterials.2010.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/14/2010] [Indexed: 12/14/2022]
Abstract
The artificial dsRNA polyriboinosinic acid-polyribocytidylic acid, poly(I:C), is a potent adjuvant candidate for vaccination, as it strongly drives cell-mediated immunity. However, because of its effects on non-immune bystander cells, poly(I:C) administration may bear danger for the development of autoimmune diseases. Thus poly(I:C) should be applied in the lowest dose possible. We investigated microspheres carrying surface-assembled poly(I:C) as a two-in-one adjuvant formulation to stimulate maturation of monocyte-derived dendritic cells (MoDCs). Negatively charged polystyrene microspheres were equipped with a poly(ethylene glycol) corona through electrostatically driven surface assembly of a library of polycationic poly(l-lysine)-graft-poly(ethylene glycol) copolymers, PLL-g-PEG. Stable surface assembly of poly(I:C) was achieved by incubation of polymer-coated microspheres in an aqueous poly(I:C) solution. Surface-assembled poly(I:C) exhibited a strongly enhanced efficacy to stimulate maturation of MoDCs by up to two orders of magnitude, as compared to free poly(I:C). Multiple phagocytosis events were the key factor to enhance the efficacy. The cytokine secretion pattern of MoDCs after exposure to surface-assembled poly(I:C) differed from that of free poly(I:C), while their ability to stimulate T cell proliferation was similar. Overall, phagocytic signaling plays an important role in defining the resulting immune response to such two-in-one adjuvant formulations.
Collapse
Affiliation(s)
- Annina M Hafner
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| | | | | | | |
Collapse
|