1
|
Cerdeira CD, Brigagão MRPL. Targeting Macrophage Polarization in Infectious Diseases: M1/M2 Functional Profiles, Immune Signaling and Microbial Virulence Factors. Immunol Invest 2024; 53:1030-1091. [PMID: 38913937 DOI: 10.1080/08820139.2024.2367682] [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] [Indexed: 06/26/2024]
Abstract
INTRODUCTION An event of increasing interest during host-pathogen interactions is the polarization of patrolling/naive monocytes (MOs) into macrophage subsets (MФs). Therapeutic strategies aimed at modulating this event are under investigation. METHODS This review focuses on the mechanisms of induction/development and profile of MФs polarized toward classically proinflammatory (M1) or alternatively anti-inflammatory (M2) phenotypes in response to bacteria, fungi, parasites, and viruses. RESULTS AND DISCUSSION It highlights nuclear, cytoplasmic, and cell surface receptors (pattern recognition receptors/PPRs), microenvironmental mediators, and immune signaling. MФs polarize into phenotypes: M1 MФs, activated by IFN-γ, pathogen-associated molecular patterns (PAMPs, e.g. lipopolysaccharide) and membrane-bound PPRs ligands (TLRs/CLRs ligands); or M2 MФs, induced by interleukins (ILs-4, -10 and -13), antigen-antibody complexes, and helminth PAMPs. Polarization toward M1 and M2 profiles evolve in a pathogen-specific manner, with or without canonicity, and can vary widely. Ultimately, this can result in varying degrees of host protection or more severe disease outcome. On the one hand, the host is driving effective MФs polarization (M1 or M2); but on the other hand, microorganisms may skew the polarization through virulence factors to increase pathogenicity. Cellular/genomic reprogramming also ensures plasticity of M1/M2 phenotypes. Because modulation of polarization can occur at multiple points, new insights and emerging perspectives may have clinical implications during the inflammation-to-resolution transition; translated into practical applications as for therapeutic/vaccine design target to boost microbicidal response (M1, e.g. triggering oxidative burst) with specifics PAMPs/IFN-γ or promote tissue repair (M2, increasing arginase activity) via immunotherapy.
Collapse
|
2
|
Duan H, Meng F, Liu X, Qi P, Peng X, Li C, Wang Q, Zhao G, Lin J. Extracellular vesicles from Candida albicans modulate immune cells function and play a protective role in fungal keratitis. Microb Pathog 2024; 189:106606. [PMID: 38437994 DOI: 10.1016/j.micpath.2024.106606] [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: 01/21/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
Fungal keratitis (FK) is a highly blinding infectious corneal disease caused by pathogenic fungi. Candida albicans (C. albicans) is one of the main pathogens of fungal keratitis. Extracellular vesicles (EVs), lipid bilayer compartments released by almost all living cells, including fungi, have garnered attention for their role in pathogenic microbial infection and host immune responses in recent years. Studies have reported that pretreating the host with fungal EVs can reduce the inflammatory response of the host when attacked by fungi and reduce the lethality of fungal infection. However, there are no studies that have evaluated whether C. albicans EVs can modulate the inflammatory response associated with C. albicans keratitis. Our study revealed that C. albicans EVs could activate the polymorphonuclear cells (PMNs) and promote their secretion of proinflammatory cytokines and nitric oxide (NO), enhance their phagocytic and fungicidal abilities against C. albicans. C. albicans EVs also induced a proinflammatory response in RAW264.7 cells, which was characterized by increased production of inflammatory cytokines and elevated expression of the chemokine CCL2. Similarly, stimulation of C. albicans EVs to RAW264.7 cells also enhanced the phagocytosis and killing ability of cells against C. albicans. Besides, in our in vivo experiments, after receiving subconjunctival injection of C. albicans EVs, C57BL/6 mice were infected with C. albicans. The results demonstrated that pre-exposure to C. albicans EVs could effectively diminish the severity of keratitis, reduce fungal load and improve prognosis. Overall, we conclude that C. albicans EVs can modulate the function of immune cells and play a protective role in C. albicans keratitis.
Collapse
Affiliation(s)
- Huijin Duan
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Fanyue Meng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xing Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Pingli Qi
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xudong Peng
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
3
|
Mattke J, Darden CM, Lawrence MC, Kuncha J, Shah YA, Kane RR, Naziruddin B. Toll-like receptor 4 in pancreatic damage and immune infiltration in acute pancreatitis. Front Immunol 2024; 15:1362727. [PMID: 38585277 PMCID: PMC10995222 DOI: 10.3389/fimmu.2024.1362727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Acute pancreatitis is a complex inflammatory disease resulting in extreme pain and can result in significant morbidity and mortality. It can be caused by several factors ranging from genetics, alcohol use, gall stones, and ductal obstruction caused by calcification or neutrophil extracellular traps. Acute pancreatitis is also characterized by immune cell infiltration of neutrophils and M1 macrophages. Toll-like receptor 4 (TLR4) is a pattern recognition receptor that has been noted to respond to endogenous ligands such as high mobility group box 1 (HMGB1) protein and or exogenous ligands such as lipopolysaccharide both of which can be present during the progression of acute pancreatitis. This receptor can be found on a variety of cell types from endothelial cells to resident and infiltrating immune cells leading to production of pro-inflammatory cytokines as well as immune cell activation and maturation resulting in the furthering of pancreatic damage during acute pancreatitis. In this review we will address the various mechanisms mediated by TLR4 in the advancement of acute pancreatitis and how targeting this receptor could lead to improved outcomes for patients suffering from this condition.
Collapse
Affiliation(s)
- Jordan Mattke
- Baylor University, Institute of Biomedical Studies, Waco, TX, United States
| | - Carly M. Darden
- Baylor University Medical Center, Annette C. and Harold C. Simmons Transplant Institute, Dallas, TX, United States
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Jayachandra Kuncha
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Yumna Ali Shah
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Robert R. Kane
- Baylor University, Institute of Biomedical Studies, Waco, TX, United States
| | - Bashoo Naziruddin
- Baylor University Medical Center, Annette C. and Harold C. Simmons Transplant Institute, Dallas, TX, United States
| |
Collapse
|
4
|
Zhang YY, Li J, Li F, Xue S, Xu QY, Zhang YQ, Feng L. Palmitic acid combined with γ-interferon inhibits gastric cancer progression by modulating tumor-associated macrophages' polarization via the TLR4 pathway. J Cancer Res Clin Oncol 2023; 149:7053-7067. [PMID: 36862159 DOI: 10.1007/s00432-023-04655-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/14/2023] [Indexed: 03/03/2023]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) constitute the main infiltrating immune cells in the solid tumor microenvironment. Amounting studies have analyzed the antitumor effect on immune response induced by Toll-like receptor (TLR) agonists, such as lipopolysaccharide (LPS), γ-interferon (γ-IFN), and palmitic Acid (PA). However, their combined treatment for gastric cancer (GC) has not been illuminated. METHODS We investigated the relevance of macrophage polarization and the effect of PA and γ-IFN in GC in vitro and in vivo. M1 and M2 macrophage-associated markers were measured by real-time quantitative PCR and flow cytometry, and the activation level of the TLR4 signaling pathways was evaluated by western blot analysis. The effect of PA and γ-IFN on the proliferation, migration, and invasion of GC cells (GCCs) was evaluated by Cell-Counting Kit-8, transwell assays, and wound-healing assays. In vivo animal models were used to verify the effect of PA and γ-IFN on tumor progression, and the M1 and M2 macrophage markers, CD8 + T lymphocytes, regulatory T cells (Treg) cells, and the myeloid-derived suppressor cells (MDSCs) in tumor tissues were analyzed by flow cytometry and immunohistochemical (IHC). RESULTS The results showed that this combination strategy enhanced M1-like macrophages and diminished M2-like macrophages through the TLR4 signaling pathway in vitro. In addition, the combination strategy impairs the proliferative and migratory activity of GCC in vitro and in vivo. While, the antitumor effect was abolished using the TAK-424 (a specific TLR-4 signaling pathway inhibitor) in vitro. CONCLUSIONS The combined treatment of PA and γ-IFN inhibited GC progression by modulating macrophages polarization via the TLR4 pathway.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Shuai Xue
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Qing-Yu Xu
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ya-Qiong Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
5
|
Freitas MS, Bitencourt TA, Rezende CP, Martins NS, Dourado TDMH, Tirapelli CR, Almeida F. Aspergillus fumigatus Extracellular Vesicles Display Increased Galleria mellonella Survival but Partial Pro-Inflammatory Response by Macrophages. J Fungi (Basel) 2023; 9:jof9050541. [PMID: 37233252 DOI: 10.3390/jof9050541] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Fungal extracellular vesicles (EVs) mediate intra- and interspecies communication and are critical in host-fungus interaction, modulating inflammation and immune responses. In this study, we evaluated the in vitro pro- and anti-inflammatory properties of Aspergillus fumigatus EVs over innate leukocytes. A. fumigatus EVs induced a partial proinflammatory response by macrophages, characterized by increased tumor necrosis factor-alpha production, and increased gene expression of induced nitric oxide synthase and adhesion molecules. EVs induce neither NETosis in human neutrophils nor cytokine secretion by peripheral mononuclear cells. However, prior inoculation of A. fumigatus EVs in Galleria mellonella larvae resulted in increased survival after the fungal challenge. Taken together, these findings show that A. fumigatus EVs play a role in protection against fungal infection, although they induce a partial pro-inflammatory response.
Collapse
Affiliation(s)
- Mateus Silveira Freitas
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Tamires Aparecida Bitencourt
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Caroline Patini Rezende
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Nubia Sabrina Martins
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | | | - Carlos R Tirapelli
- Laboratory of Pharmacology, Department of Psychiatric Nursing and Human Sciences, College of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-902, SP, Brazil
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| |
Collapse
|
6
|
Licá ICL, Frazão GCCG, Nogueira RA, Lira MGS, dos Santos VAF, Rodrigues JGM, Miranda GS, Carvalho RC, Silva LA, Guerra RNM, Nascimento FRF. Immunological mechanisms involved in macrophage activation and polarization in schistosomiasis. Parasitology 2023; 150:401-415. [PMID: 36601859 PMCID: PMC10089811 DOI: 10.1017/s0031182023000021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
Human schistosomiasis is caused by helminths of the genus Schistosoma. Macrophages play a crucial role in the immune regulation of this disease. These cells acquire different phenotypes depending on the type of stimulus they receive. M1 macrophages can be ‘classically activated’ and can display a proinflammatory phenotype. M2 or ‘alternatively activated’ macrophages are considered anti-inflammatory cells. Despite the relevance of macrophages in controlling infections, the role of the functional types of these cells in schistosomiasis is unclear. This review highlights different molecules and/or macrophage activation and polarization pathways during Schistosoma mansoni and Schistosoma japonicum infection. This review is based on original and review articles obtained through searches in major databases, including Scopus, Google Scholar, ACS, PubMed, Wiley, Scielo, Web of Science, LILACS and ScienceDirect. Our findings emphasize the importance of S. mansoni and S. japonicum antigens in macrophage polarization, as they exert immunomodulatory effects in different stages of the disease and are therefore important as therapeutic targets for schistosomiasis and in vaccine development. A combination of different antigens can provide greater protection, as it possibly stimulates an adequate immune response for an M1 or M2 profile and leads to host resistance; however, this warrants in vitro and in vivo studies.
Collapse
Affiliation(s)
- Irlla Correia Lima Licá
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Gleycka Cristine Carvalho Gomes Frazão
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Ranielly Araujo Nogueira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Maria Gabriela Sampaio Lira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Vitor Augusto Ferreira dos Santos
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - João Gustavo Mendes Rodrigues
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Guilherme Silva Miranda
- Department of Biology, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Lucilene Amorim Silva
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Rosane Nassar Meireles Guerra
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Flávia Raquel Fernandes Nascimento
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| |
Collapse
|
7
|
Chi G, Pei J, Li X. The imbalance of liver resident macrophages polarization promotes chronic autoimmune hepatitis development in mice. PeerJ 2023; 11:e14871. [PMID: 36778150 PMCID: PMC9912947 DOI: 10.7717/peerj.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Background Autoimmune hepatitis (AIH) is a chronic immune-mediated inflammatory liver disease. At present, it is largely unknown how the innate immune cells influence AIH development. Objective To inquiry about mechanism of liver resident macrophages in AIH development, thus offering a new direction for AIH targeted treatment. Methods The liver resident macrophages were eliminated by clodronate liposomes in AIH liver tissues, followed by HE and Picrosirius assay to detect liver fibrosis and lymphocyte infiltration. The liver resident macrophages polarization was detected by Immunohistochemistry and qPCR. The collagenase digestion was used to isolate Kupffer cells from AIH mice liver tissues and pro-/anti-inflammatory cytokines were determined by qPCR. Results M2 macrophages were the dominant phenotype at early immune response stage and hepatic inflammation was progressively aggravated after depletion of liver resident macrophages. M2 macrophages could effectively delay the development of AIH and could be polarized to M1 macrophages at the disease progresses. TLR2 ligands could promote M2 macrophages producing anti-inflammatory cytokines, whereas TLR4 ligands could promote M1 macrophages producing proinflammatory cytokines. The change of TLR2 and TLR4 ligands could lead to continuous high expression of TLR4 and decreased expression of TLR2 in macrophages to further affect liver resident macrophages polarization state. Conclusion TLR2 and TLR4 ligands mediated liver resident macrophages polarization to favor chronic autoimmune hepatitis development.
Collapse
|
8
|
Silva MJA, Santana DS, de Oliveira LG, Monteiro EOL, Lima LNGC. The relationship between 896A/G (rs4986790) polymorphism of TLR4 and infectious diseases: A meta-analysis. Front Genet 2022; 13:1045725. [PMID: 36506333 PMCID: PMC9729345 DOI: 10.3389/fgene.2022.1045725] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
Toll-like Receptors (TLRs), such as the TLR4, are genes encoding transmembrane receptors of the same name, which induce a pro- or anti-inflammatory response according to their expression as the host's first line of defense against pathogens, such as infectious ones. Single nucleotide polymorphisms (SNPs) are the most common type of mutation in the human genome and can generate functional modification in genes. The aim of this article is to review in which infectious diseases there is an association of susceptibility or protection by the TLR4 SNP rs4986790. A systematic review and meta-analysis of the literature was conducted in the Science Direct, PUBMED, MEDLINE, and SciELO databases between 2011 and 2021 based on the dominant genotypic model of this SNP for general and subgroup analysis of infectious agent type in random effect. Summary odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were calculated for genotypic comparison. I2 statistics were calculated to assess the presence of heterogeneity between studies and funnel plots were inspected for indication of publication bias. A total of 27 articles were included, all in English. Among the results achieved, the categories of diseases that were most associated with the SNP studied were in decreasing order of number of articles: infections by bacteria (29.63%); caused by viruses (22.23%); urinary tract infection-UTI (7.4%), while 11 studies (40.74%) demonstrated a nonsignificant association. In this meta-analysis, a total of 5599 cases and 5871 controls were finalized. The present meta-analysis suggests that there is no significant association between TLR4-rs4986790 SNP and infections (OR = 1,11; 95% CI: 0,75-1,66; p = 0,59), but in the virus subgroup it was associated with a higher risk (OR = 2,16; 95% CI: 1,09-4,30; p = 0,03). The subgroups of bacteria and parasites did not show statistical significance (OR = 0,86; 95% CI: 0,56-1,30; p = 0,47, and no estimate of effects, respectively). Therefore, it has been shown that a diversity of infectious diseases is related to this polymorphism, either by susceptibility or even severity to them, and the receptor generated is also crucial for the generation of cell signaling pathways and immune response against pathogens.
Collapse
Affiliation(s)
| | - Davi Silva Santana
- Institute of Health Sciences (ICS), Federal University of Pará (UFPA), Belém, Brazil
| | | | | | | |
Collapse
|
9
|
Proteoglycan 4 (PRG4) treatment enhances wound closure and tissue regeneration. NPJ Regen Med 2022; 7:32. [PMID: 35750773 PMCID: PMC9232611 DOI: 10.1038/s41536-022-00228-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 05/20/2022] [Indexed: 01/13/2023] Open
Abstract
The wound healing response is one of most primitive and conserved physiological responses in the animal kingdom, as restoring tissue integrity/homeostasis can be the difference between life and death. Wound healing in mammals is mediated by immune cells and inflammatory signaling molecules that regulate tissue resident cells, including local progenitor cells, to mediate closure of the wound through formation of a scar. Proteoglycan 4 (PRG4), a protein found throughout the animal kingdom from fish to elephants, is best known as a glycoprotein that reduces friction between articulating surfaces (e.g. cartilage). Previously, PRG4 was also shown to regulate the inflammatory and fibrotic response. Based on this, we asked whether PRG4 plays a role in the wound healing response. Using an ear wound model, topical application of exogenous recombinant human (rh)PRG4 hastened wound closure and enhanced tissue regeneration. Our results also suggest that rhPRG4 may impact the fibrotic response, angiogenesis/blood flow to the injury site, macrophage inflammatory dynamics, recruitment of immune and increased proliferation of adult mesenchymal progenitor cells (MPCs) and promoting chondrogenic differentiation of MPCs to form the auricular cartilage scaffold of the injured ear. These results suggest that PRG4 has the potential to suppress scar formation while enhancing connective tissue regeneration post-injury by modulating aspects of each wound healing stage (blood clotting, inflammation, tissue generation and tissue remodeling). Therefore, we propose that rhPRG4 may represent a potential therapy to mitigate scar and improve wound healing.
Collapse
|
10
|
Pitangui NDS, Fernandes FF, Gonçales RA, Roque-Barreira MC. Virulence Vs. Immunomodulation: Roles of the Paracoccin Chitinase and Carbohydrate-Binding Sites in Paracoccidioides brasiliensis Infection. Front Mol Biosci 2021; 8:700797. [PMID: 34532342 PMCID: PMC8438136 DOI: 10.3389/fmolb.2021.700797] [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: 04/26/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
Paracoccin (PCN) is a bifunctional protein primarily present in the cell wall of Paracoccidioides brasiliensis, a human pathogenic dimorphic fungus. PCN has one chitinase region and four potential lectin sites and acts as both a fungal virulence factor and an immunomodulator of the host response. The PCN activity on fungal virulence, mediated by the chitinase site, was discovered by infecting mice with yeast overexpressing PCN (PCN-ov). PCN-ov are characterized by increased chitin hydrolysis, a narrow cell wall, and augmented resistance to phagocytes' fungicidal activity. Compared to wild-type (wt) yeast, infection with PCN-ov yeast causes a more severe disease, which is attributed to the increased PCN chitinase activity. In turn, immunomodulation of the host response was demonstrated by injecting, subcutaneously, recombinant PCN in mice infected with wt-P. brasiliensis. Through its carbohydrate binding site, the injected recombinant PCN interacts with Toll-like receptor 2 (TLR2) and Toll-like receptor 4 (TLR4) N-glycans on macrophages, triggers M1 polarization, and stimulates protective Th1 immunity against the fungus. The PCN-treatment of wt yeast-infected mice results in mild paracoccidioidomycosis. Therefore, PCN paradoxically influences the course of murine paracoccidioidomycosis. The disease is severe when caused by yeast that overexpress endogenous PCN, which exerts a robust local chitinase activity, followed by architectural changes of the cell wall and release of low size chito-oligomers. However, the disease is mild when exogenous PCN is injected, which recognizes N-glycans on systemic macrophages resulting in immunomodulation.
Collapse
Affiliation(s)
- Nayla de Souza Pitangui
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fabrício Freitas Fernandes
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Relber Aguiar Gonçales
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
11
|
The Role of IL-17-Producing Cells in Cutaneous Fungal Infections. Int J Mol Sci 2021; 22:ijms22115794. [PMID: 34071562 PMCID: PMC8198319 DOI: 10.3390/ijms22115794] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/15/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is the outermost layer of the body and is exposed to many environmental stimuli, which cause various inflammatory immune responses in the skin. Among them, fungi are common microorganisms that colonize the skin and cause cutaneous fungal diseases such as candidiasis and dermatophytosis. The skin exerts inflammatory responses to eliminate these fungi through the cooperation of skin-component immune cells. IL-17 producing cells are representative immune cells that play a vital role in anti-fungal action in the skin by producing antimicrobial peptides and facilitating neutrophil infiltration. However, the actual impact of IL-17-producing cells in cutaneous fungal infections remains unclear. In this review, we focused on the role of IL-17-producing cells in a series of cutaneous fungal infections, the characteristics of skin infectious fungi, and the recognition of cell components that drive cutaneous immune cells.
Collapse
|
12
|
Puccia R. Current Status on Extracellular Vesicles from the Dimorphic Pathogenic Species of Paracoccidioides. Curr Top Microbiol Immunol 2021; 432:19-33. [DOI: 10.1007/978-3-030-83391-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Gonçales RA, Ricci-Azevedo R, Vieira VCS, Fernandes FF, Thomaz SMDO, Carvalho A, Vendruscolo PE, Cunha C, Roque-Barreira MC, Rodrigues F. Paracoccin Overexpression in Paracoccidioides brasiliensis Enhances Fungal Virulence by Remodeling Chitin Properties of the Cell Wall. J Infect Dis 2020; 224:164-174. [PMID: 33201217 DOI: 10.1093/infdis/jiaa707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The thermodimorphic fungi Paracoccidioides spp. are the etiological agents of paracoccidioidomycosis. Although poorly studied, paracoccin (PCN) from Paracoccidioides brasiliensis has been shown to harbor lectinic, enzymatic, and immunomodulatory properties that affect disease development. METHODS Mutants of P. brasiliensis overexpressing PCN (ov-PCN) were constructed by Agrobacterium tumefaciens-mediated transformation. ov-PCN strains were analyzed and inoculated intranasally or intravenously to mice. Fungal burden, lung pathology, and survival were monitored to evaluate virulence. Electron microscopy was used to evaluate the size of chito-oligomer particles released by ov-PCN or wild-type strains to growth media. RESULTS ov-PCN strains revealed no differences in cell growth and viability, although PCN overexpression favored cell separation, chitin processing that results in the release of smaller chito-oligomer particles, and enhanced virulence. Our data show that PCN triggers a critical effect in the cell wall biogenesis through the chitinase activity resulting from overexpression of PCN. As such, PCN overexpression aggravates the disease caused by P. brasiliensis. CONCLUSIONS Our data are consistent with a model in which PCN modulates the cell wall architecture via its chitinase activity. These findings highlight the potential for exploiting PCN function in future therapeutic approaches.
Collapse
Affiliation(s)
- Relber Aguiar Gonçales
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil.,Life and Health Sciences Research Institute School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafael Ricci-Azevedo
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil
| | - Vanessa C S Vieira
- Life and Health Sciences Research Institute School of Medicine, University of Minho, Braga, Portugal
| | - Fabrício F Fernandes
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil
| | - Sandra M de O Thomaz
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia E Vendruscolo
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil
| | - Cristina Cunha
- Life and Health Sciences Research Institute School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maria Cristina Roque-Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto/São Paulo, Brazil
| | - Fernando Rodrigues
- Life and Health Sciences Research Institute School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
14
|
Hsu LW, Huang KT, Nakano T, Chiu KW, Chen KD, Goto S, Chen CL. MicroRNA-301a inhibition enhances the immunomodulatory functions of adipose-derived mesenchymal stem cells by induction of macrophage M2 polarization. Int J Immunopathol Pharmacol 2020; 34:2058738420966092. [PMID: 33121303 PMCID: PMC7607751 DOI: 10.1177/2058738420966092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of short non-coding RNAs that play a significant role in biological processes in various cell types, including mesenchymal stem cells (MSCs). However, how miRNAs regulate the immunomodulatory functions of adipose-derived MSCs (AD-MSCs) remains unknown. Here, we showed that modulation of miR-301a in AD-MSCs altered macrophage polarization. Bone marrow (BM)-derived macrophages were stimulated with LPS (1 μg/ml) and co-cultured with miRNA transfected AD-MSCs for 24 h. The expression of M1 and M2 markers in macrophages was analyzed. Inhibition of miR-301a induced M2 macrophage with arginase-1, CD163, CD206, and IL-10 upregulation. Additionally, toll-like receptor (TLR)-4 mRNA expression in macrophages was downregulated in co-cultures with AD-MSCs transfected with a miR-301a inhibitor. Nitric oxide (NO) in the supernatant of AD-MSC/macrophage co-culture was also suppressed by inhibition of miR-301a in AD-MSCs. We further found that suppression of miR-301a in AD-MSCs increased prostaglandin E2 (PGE2) concentration in the conditioned medium of the co-culture. Taken together, the results of our study indicate that miR-301a can modulate the immunoregulatory functions of AD-MSCs that favor the applicability as a potential immunotherapeutic agent.
Collapse
Affiliation(s)
- Li-Wen Hsu
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung
| | - Kuang-Tzu Huang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung
| | - Toshiaki Nakano
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung
| | - King-Wah Chiu
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung
| | - Kuang-Den Chen
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung
| | - Shigeru Goto
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung.,Faculty of Nursing, Department of Nursing, Josai International University, Togane, Chiba, Japan
| | - Chao-Long Chen
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung
| |
Collapse
|
15
|
Unraveling the susceptibility of paracoccidioidomycosis: Insights towards the pathogen-immune interplay and immunogenetics. INFECTION GENETICS AND EVOLUTION 2020; 86:104586. [PMID: 33039601 DOI: 10.1016/j.meegid.2020.104586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Paracoccidioidomycosis (PCM) is a life-threatening systemic mycosis caused by Paracoccidioides spp. This disease comprises three clinical forms: symptomatic acute and chronic forms (PCM disease) and PCM infection, a latent form without clinical symptoms. PCM disease differs markedly according to severity, clinical manifestations, and host immune response. Fungal virulence factors and adhesion molecules are determinants for entry, latency, immune escape and invasion, and dissemination in the host. Neutrophils and macrophages play a paramount role in first-line defense against the fungus through the recognition of antigens by pattern recognition receptors (PRRs), activating their microbicidal machinery. Furthermore, the clinical outcome of the PCM is strongly associated with the variability of cytokines and immunoglobulins produced by T and B cells. While the mechanisms that mediate susceptibility or resistance to infection are dictated by the immune system, some genetic factors may alter gene expression and its final products and, hence, modulate how the organism responds to infection and injury. This review outlines the main findings relative to this topic, addressing the complexity of the immune response triggered by Paracoccidioides spp. infection from preclinical investigations to studies in humans. Here, we focus on mechanisms of fungal pathogenesis, the patterns of innate and adaptive immunity, and the genetic and molecular basis related to immune response and susceptibility to the development of the PCM and its clinical forms. Immunogenetic features such as HLA system, cytokines/cytokines receptors genes and other immune-related genes, and miRNAs are likewise discussed. Finally, we point out the occurrence of PCM in patients with primary immunodeficiencies and call attention to the research gaps and challenges faced by the PCM field.
Collapse
|
16
|
Feng X, Yu W, Cao L, Meng F, Cong M. A novel chrysin thiazole derivative polarizes macrophages to an M1 phenotype via targeting TLR4. Int Immunopharmacol 2020; 88:106986. [PMID: 33182070 DOI: 10.1016/j.intimp.2020.106986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) are an important cause of tumorigenesis and tumor development. M2 macrophages can promote tumor growth while M1 macrophages kill tumor cells, therefore, polarizing macrophages to achieve a functional M1 phenotype could effectively play its anti-tumor role. In the current study, we synthesized a novel chrysin derivative which is termed as ChR-TD. And we found ChR-TD might be a ligand of TLR4 that polarized the TAMs towards M1 phenotype and played its anti-tumor role. Further study indicated that ChR-TD reprogrammed the macrophages into an M1 phenotype via TLR4 activation. Moreover, ChR-TD activated TLR4/NF-κB signaling pathway and promoted the NF-κB/p65 translocated into the nuclear, leading to the activation of NF-κB and proinflammatory cytokines release. In addition, type I interferon signaling was also activated by ChR-TD, leading to the expressions of IFN-α and IFN-β and its targeted genes NOS2, MCP-1 and IP-10 were significantly increased in macrophages. Importantly, these effects were disturbed in TLR4-/- macrophages, which are constructed by using CRISPR/Cas9 system. And the molecule docking simulation further indicated that ChR-TD could bind to TLR4 and might be a ligand of TLR4. Hence, these findings suggested that ChR-TD might be a ligand of TLR4 and can be used as a potential lead compound for tumors treatment.
Collapse
Affiliation(s)
- Xiujing Feng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China.
| | - Wen Yu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210046, China
| | - Lingsen Cao
- Key Lab of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Fanda Meng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| | - Mulin Cong
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| |
Collapse
|
17
|
Abstract
Immunocompromised patients are susceptible to several fungal infections. The genus Aspergillus can cause increased morbidity and mortality. Developing new therapies is essential to understand the fungal biology mechanisms. Fungal EVs carry important virulence factors, thus playing pivotal roles in fungal pathophysiology. No study to date has reported EV production by Aspergillus flavus, a fungus considered to be the second most common cause of aspergillosis and relevant food contaminator found worldwide. In this study, we produced A. flavus EVs and evaluated the in vitro immunomodulatory effects of EVs on bone marrow-derived macrophages (BMDMs) and in vivo effects in a Galleria mellonella model. Aspergillus flavus, a ubiquitous and saprophytic fungus, is the second most common cause of aspergillosis worldwide. Several mechanisms contribute to the establishment of the fungal infection. Extracellular vesicles (EVs) have been described as “virulence factor delivery bags” in several fungal species, demonstrating a crucial role during the infection. In this study, we evaluated production of A. flavus EVs and their immunomodulatory functions. We verified that A. flavus EVs induce macrophages to produce inflammatory mediators, such as nitric oxide, tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and IL-1β. Furthermore, the A. flavus EVs enhance phagocytosis and killing by macrophages and induce M1 macrophage polarization in vitro. In addition, a prior inoculation of A. flavus EVs in Galleria mellonella larvae resulted in a protective effect against the fungal infection. Our findings suggest that A. flavus EVs are biologically active and affect the interaction between A. flavus and host immune cells, priming the innate immune system to eliminate the fungal infection. Collectively, our results suggest that A. flavus EVs play a crucial role in aspergillosis. IMPORTANCE Immunocompromised patients are susceptible to several fungal infections. The genus Aspergillus can cause increased morbidity and mortality. Developing new therapies is essential to understand the fungal biology mechanisms. Fungal EVs carry important virulence factors, thus playing pivotal roles in fungal pathophysiology. No study to date has reported EV production by Aspergillus flavus, a fungus considered to be the second most common cause of aspergillosis and relevant food contaminator found worldwide. In this study, we produced A. flavus EVs and evaluated the in vitro immunomodulatory effects of EVs on bone marrow-derived macrophages (BMDMs) and in vivo effects in a Galleria mellonella model.
Collapse
|
18
|
Oliveira-Brito PKM, Rezende CP, Almeida F, Roque-Barreira MC, da Silva TA. iNOS/Arginase-1 expression in the pulmonary tissue over time during Cryptococcus gattii infection. Innate Immun 2019; 26:117-129. [PMID: 31446837 PMCID: PMC7016403 DOI: 10.1177/1753425919869436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Inhalation of Cryptococcus gattii yeasts (causing cryptococcosis) triggers an anti-cryptococcal immune response initiated by macrophages, neutrophils or dendritic cells, and the iNOS expressed by various cells may regulate the function and differentiation of innate and adaptive immune cells. Here, we evaluated the effect of progression of C. gattii infection on the host innate immune response. C. gattii infection in BALB/c mice spreads to several organs by 21 d post infection. The numbers of neutrophils and lymphocytes in the peripheral blood of C. gattii–infected mice were remarkably altered on that day. The frequency of CD11b+ cells and cell concentrations of CD4+ and CD8+ T cells was significantly altered in the pulmonary tissue of infected mice. We found a higher frequency of CD11b+/iNOS+ cells in the lungs of infected mice, accompanied by an increase in frequency of CD11b+/Arginase-1+ cells over time. Moreover, the iNOS/Arginase-1 expression ratio in CD11b+ cells reached its lowest value at 21 d post infection. In addition, the cytokine micro-environment in infected lungs did not show a pro-inflammatory profile. Surprisingly, iNOS knock-out prolonged the survival of infected mice, while their pulmonary fungal burden was higher than that of infected WT mice. Thus, C. gattii infection alters the immune response in the pulmonary tissue, and iNOS expression may play a key role in infection progression.
Collapse
Affiliation(s)
- Patrícia Kellen Martins Oliveira-Brito
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil.,These authors contributed equally to this work
| | - Caroline Patini Rezende
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Brazil.,These authors contributed equally to this work
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Thiago Aparecido da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Brazil
| |
Collapse
|
19
|
The lectin-specific activity of Toxoplasma gondii microneme proteins 1 and 4 binds Toll-like receptor 2 and 4 N-glycans to regulate innate immune priming. PLoS Pathog 2019; 15:e1007871. [PMID: 31226171 PMCID: PMC6608980 DOI: 10.1371/journal.ppat.1007871] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/03/2019] [Accepted: 05/25/2019] [Indexed: 01/01/2023] Open
Abstract
Infection of host cells by Toxoplasma gondii is an active process, which is regulated by secretion of microneme (MICs) and rhoptry proteins (ROPs and RONs) from specialized organelles in the apical pole of the parasite. MIC1, MIC4 and MIC6 assemble into an adhesin complex secreted on the parasite surface that functions to promote infection competency. MIC1 and MIC4 are known to bind terminal sialic acid residues and galactose residues, respectively and to induce IL-12 production from splenocytes. Here we show that rMIC1- and rMIC4-stimulated dendritic cells and macrophages produce proinflammatory cytokines, and they do so by engaging TLR2 and TLR4. This process depends on sugar recognition, since point mutations in the carbohydrate-recognition domains (CRD) of rMIC1 and rMIC4 inhibit innate immune cells activation. HEK cells transfected with TLR2 glycomutants were selectively unresponsive to MICs. Following in vitro infection, parasites lacking MIC1 or MIC4, as well as expressing MIC proteins with point mutations in their CRD, failed to induce wild-type (WT) levels of IL-12 secretion by innate immune cells. However, only MIC1 was shown to impact systemic levels of IL-12 and IFN-γ in vivo. Together, our data show that MIC1 and MIC4 interact physically with TLR2 and TLR4 N-glycans to trigger IL-12 responses, and MIC1 is playing a significant role in vivo by altering T. gondii infection competency and murine pathogenesis. Toxoplasmosis is caused by the protozoan Toxoplasma gondii, belonging to the Apicomplexa phylum. This phylum comprises important parasites able to infect a broad diversity of animals, including humans. A particularity of T. gondii is its ability to invade virtually any nucleated cell of all warm-blooded animals through an active process, which depends on the secretion of adhesin proteins. These proteins are discharged by specialized organelles localized in the parasite apical region, and termed micronemes and rhoptries. We show in this study that two microneme proteins from T. gondii utilize their adhesion activity to stimulate innate immunity. These microneme proteins, denoted MIC1 and MIC4, recognize specific sugars on receptors expressed on the surface of mammalian immune cells. This binding activates these innate immune cells to secrete cytokines, which promotes efficient host defense mechanisms against the parasite and regulate their pathogenesis. This activity promotes a chronic infection by controlling parasite replication during acute infection.
Collapse
|
20
|
Bai D, Zhao Y, Zhu Q, Zhou Y, Zhao Y, Zhang T, Guo Q, Lu N. LZ205, a newly synthesized flavonoid compound, exerts anti-inflammatory effect by inhibiting M1 macrophage polarization through regulating PI3K/AKT/mTOR signaling pathway. Exp Cell Res 2018; 364:84-94. [DOI: 10.1016/j.yexcr.2018.01.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/20/2018] [Accepted: 01/24/2018] [Indexed: 01/26/2023]
|
21
|
Schäfer C, Ascui G, Ribeiro CH, López M, Prados-Rosales R, González PA, Bueno SM, Riedel CA, Baena A, Kalergis AM, Carreño LJ. Innate immune cells for immunotherapy of autoimmune and cancer disorders. Int Rev Immunol 2017; 36:315-337. [PMID: 28933579 DOI: 10.1080/08830185.2017.1365145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Modulation of the immune system has been widely targeted for the treatment of several immune-related diseases, such as autoimmune disorders and cancer, due to its crucial role in these pathologies. Current available therapies focus mainly on symptomatic treatment and are often associated with undesirable secondary effects. For several years, remission of disease and subsequently recovery of immune homeostasis has been a major goal for immunotherapy. Most current immunotherapeutic strategies are aimed to inhibit or potentiate directly the adaptive immune response by modulating antibody production and B cell memory, as well as the effector potential and memory of T cells. Although these immunomodulatory approaches have shown some success in the clinic with promising therapeutic potential, they have some limitations related to their effectiveness in disease models and clinical trials, as well as elevated costs. In the recent years, a renewed interest has emerged on targeting innate immune cells for immunotherapy, due to their high plasticity and ability to exert a potent and extremely rapid response, which can influence the outcome of the adaptive immune response. In this review, we discuss the immunomodulatory potential of several innate immune cells, as well as they use for immunotherapy, especially in autoimmunity and cancer.
Collapse
Affiliation(s)
- Carolina Schäfer
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Gabriel Ascui
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Carolina H Ribeiro
- b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Mercedes López
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| | - Rafael Prados-Rosales
- c Centro de Investigaciones Cooperativas en Biociencias (CIC bioGUNE) , Bilbao , Spain
| | - Pablo A González
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Susan M Bueno
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Claudia A Riedel
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,e Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina , Universidad Andrés Bello , Santiago , Chile
| | - Andrés Baena
- f Departamento de Microbiología y Parasitología, Facultad de Medicina , Universidad de Antioquia , Medellín , Colombia
| | - Alexis M Kalergis
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,d Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile.,g Departamento de Endocrinología, Facultad de Medicina , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Leandro J Carreño
- a Millennium Institute on Immunology and Immunotherapy Santiago , Chile.,b Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina , Universidad de Chile , Santiago , Chile
| |
Collapse
|
22
|
Landgraf TN, Costa MV, Oliveira AF, Ribeiro WC, Panunto-Castelo A, Fernandes FF. Involvement of Dihydrolipoyl Dehydrogenase in the Phagocytosis and Killing of Paracoccidioides brasiliensis by Macrophages. Front Microbiol 2017; 8:1803. [PMID: 28970825 PMCID: PMC5609111 DOI: 10.3389/fmicb.2017.01803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/05/2017] [Indexed: 01/15/2023] Open
Abstract
Paracoccidioides brasiliensis and Paracoccidioides lutzii are fungi causing paracoccidioidomycosis (PCM), an autochthonous systemic mycosis found in Latin America. These microorganisms contain a multitude of molecules that may be associated with the complex interaction of the fungus with the host. Here, we identify the enzyme dihydrolipoyl dehydrogenase (DLD) as an exoantigen from P. brasiliensis (Pb18_Dld) by mass spectrometry. Interestingly, the DLD gene expression in yeast form showed higher expression levels than those in mycelial form and transitional phases. Pb18_Dld gene was cloned, and the recombinant protein (rPb18_Dld) was expressed and purified for subsequent studies and production of antibodies. Immunogold labeling and transmission electron microscopy revealed that the Pb18_Dld is also localized in mitochondria and cytoplasm of P. brasiliensis. Moreover, when macrophages were stimulated with rPb18Dld, there was an increase in the phagocytic and microbicidal activity of these cells, as compared with non-stimulated cells. These findings suggest that Pb18_Dld can be involved in the pathogen-host interaction, opening possibilities for studies of this protein in PCM.
Collapse
Affiliation(s)
- Taise N Landgraf
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil
| | - Marcelo V Costa
- Department of Biology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Aline F Oliveira
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil
| | - Wander C Ribeiro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil
| | - Ademilson Panunto-Castelo
- Department of Biology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Fabrício F Fernandes
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil
| |
Collapse
|
23
|
Abstract
Among the endemic deep mycoses in Latin America, paracoccidioidomycosis (PCM), caused by thermodimorphic fungi of the Paracoccidioides genus, is a major cause of morbidity. Disease development and its manifestations are associated with both host and fungal factors. Concerning the latter, several recent studies have employed the methodology of gene modulation in P. brasiliensis using antisense RNA (AsRNA) and Agrobacterium tumefaciens-mediated transformation (ATMT) to identify proteins that influence fungus virulence. Our previous observations suggested that paracoccin (PCN), a multidomain fungal protein with both lectin and enzymatic activities, may be a potential P. brasiliensis virulence factor. To explore this, we used AsRNA and ATMT methodology to obtain three independent PCN-silenced P. brasiliensis yeast strains (AsPCN1, AsPCN2, and AsPCN3) and characterized them with regard to P. brasiliensis biology and pathogenicity. AsPCN1, AsPCN2, and AsPCN3 showed relative PCN expression levels that were 60%, 40%, and 60% of that of the wild-type (WT) strain, respectively. PCN silencing led to the aggregation of fungal cells, blocked the morphological yeast-to-mycelium transition, and rendered the yeast less resistant to macrophage fungicidal activity. In addition, mice infected with AsPCN1, AsPCN2, and AsPCN3 showed a reduction in fungal burden of approximately 96% compared with those inoculated with the WT strain, which displayed a more extensive destruction of lung tissue. Finally, mice infected with the PCN-silenced yeast strains had lower mortality than those infected with the WT strain. These data demonstrate that PCN acts as a P. brasiliensis contributory virulence factor directly affecting fungal pathogenesis. The nonexistence of efficient genetic transformation systems has hampered studies in the dimorphic fungus Paracoccidioides brasiliensis, the etiological agent of the most frequent systemic mycosis in Latin America. The recent development of a method for gene expression knockdown by antisense RNA technology, associated with an Agrobacterium tumefaciens-mediated transformation system, provides new strategies for studying P. brasiliensis. Through this technology, we generated yeasts that were silenced for paracoccin (PCN), a P. brasiliensis component that has lectin and enzymatic properties. By comparing the phenotypes of PCN-silenced and wild-type strains of P. brasiliensis, we identified PCN as a virulence factor whose absence renders the yeasts unable to undergo the transition to mycelium and causes a milder pulmonary disease in mice, with a lower mortality rate. Our report highlights the importance of the technology used for P. brasiliensis transformation and demonstrates that paracoccin is a virulence factor acting on fungal biology and pathogenesis.
Collapse
|
24
|
Ramos ELP, Santana SS, Silva MV, Santiago FM, Mineo TWP, Mineo JR. Lectins from Synadenium carinatum (ScLL) and Artocarpus heterophyllus (ArtinM) Are Able to Induce Beneficial Immunomodulatory Effects in a Murine Model for Treatment of Toxoplasma gondii Infection. Front Cell Infect Microbiol 2016; 6:164. [PMID: 27933277 PMCID: PMC5122570 DOI: 10.3389/fcimb.2016.00164] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/11/2016] [Indexed: 01/09/2023] Open
Abstract
Infection by Toxoplasma gondii affects around one-third of world population and the treatment for patients presenting toxoplasmosis clinically manifested disease is mainly based by a combination of sulfadiazine, pyrimethamine, and folinic acid. However, this therapeutic protocol is significantly toxic, causing relevant dose-related bone marrow damage. Thus, it is necessary to improve new approaches to investigate the usefulness of more effective and non-toxic agents for treatment of patients with toxoplasmosis. It has been described that lectins from plants can control parasite infections, when used as immunological adjuvants in vaccination procedures. This type of lectins, such as ArtinM and ScLL is able to induce immunostimulatory activities, including efficient immune response against parasites. The present study aimed to evaluate the potential immunostimulatory effect of ScLL and ArtinM for treatment of T. gondii infection during acute phase, considering that there is no study in the literature accomplishing this issue. For this purpose, bone marrow-derived macrophages (BMDMs) were treated with different concentrations from each lectin to determine the maximum concentration without or with lowest cytotoxic effect. After, it was also measured the cytokine levels produced by these cells when stimulated by the selected concentrations of lectins. We found that ScLL showed high capacity to induce of pro-inflammatory cytokine production, while ArtinM was able to induce especially an anti-inflammatory cytokines production. Furthermore, both lectins were able to increase NO levels. Next, we evaluated the treatment effect of ScLL and ArtinM in C57BL/6 mice infected by ME49 strain from T. gondii. The animals were infected and treated with ScLL, ArtinM, ArtinM plus ScLL, or sulfadiazine, and the following parameters analyzed: Cytokines production, brain parasite burden and survival rates. Our results demonstrated that the ScLL or ScLL plus ArtinM treatment induced production of pro-inflammatory and anti-inflammatory cytokines, showing differential but complementary profiles. Moreover, when compared with non-treated mice, the parasite burden was significantly lower and survival rates higher in mice treated with ScLL or ScLL plus ArtinM, similarly with sulfadiazine treatment. In conclusion, the results demonstrated the suitable potential immunotherapeutic effect of ScLL and ArtinM lectins to control acute toxoplasmosis in this experimental murine model.
Collapse
Affiliation(s)
| | - Eliézer L P Ramos
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Silas S Santana
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Murilo V Silva
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Fernanda M Santiago
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| | - Tiago W P Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| | - José R Mineo
- Laboratório de Imunoparasitologia, Instituto de Ciências Biomédicas-Universidade Federal de Uberlândia Uberlândia, Brazil
| |
Collapse
|
25
|
Extracellular vesicles from Paracoccidioides brasiliensis induced M1 polarization in vitro. Sci Rep 2016; 6:35867. [PMID: 27775058 PMCID: PMC5075875 DOI: 10.1038/srep35867] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/07/2016] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) released by eukaryotes, archaea, and bacteria contain proteins, lipids, polysaccharides, and other molecules. The cargo analysis of EVs shows that they contain virulence factors suggesting a role in the pathogenesis of infection. The proteome, lipidome, RNA content, and carbohydrate composition of EVs from Paracoccidioides brasiliensis and Paracoccidioides lutzii were characterized. However, the effects of P. brasiliensis EVs on the host immune system have not yet been investigated. Herein, we verified that EVs from P. brasiliensis induce the production of proinflammatory mediators by murine macrophages in a dose-dependent manner. Addition of EV to macrophages also promoted transcription of the M1-polarization marker iNOs and diminish that of the M2 markers Arginase-1, Ym-1, and FIZZ-1. Furthermore, the augmented expression of M2-polarization markers, stimulated by IL-4 plus IL-10, was reverted toward an M1 phenotype in response to secondary stimulation with EVs from P. brasiliensis. The ability of EVs from P. brasiliensis to promote M1 polarization macrophages favoring an enhanced fungicidal activity, demonstrated by the decreased CFU recovery of internalized yeasts, with comparable phagocytic efficacy. Our results suggest that EVs from P. brasiliensis can modulate the innate immune response and affect the relationship between P. brasiliensis and host immune cells.
Collapse
|