1
|
Jensen O, Trujillo E, Hanson L, Ost KS. Controlling Candida: immune regulation of commensal fungi in the gut. Infect Immun 2024; 92:e0051623. [PMID: 38647290 PMCID: PMC11385159 DOI: 10.1128/iai.00516-23] [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] [Indexed: 04/25/2024] Open
Abstract
The intestinal microbiome harbors fungi that pose a significant risk to human health as opportunistic pathogens and drivers of inflammation. Inflammatory and autoimmune diseases are associated with dysbiotic fungal communities and the expansion of potentially pathogenic fungi. The gut is also the main reservoir for disseminated fungal infections. Immune interactions are critical for preventing commensal fungi from becoming pathogenic. Significant strides have been made in defining innate and adaptive immune pathways that regulate intestinal fungi, and these discoveries have coincided with advancements in our understanding of the fungal molecular pathways and effectors involved in both commensal colonization and pathogenesis within the gut. In this review, we will discuss immune interactions important for regulating commensal fungi, with a focus on how specific cell types and effectors interact with fungi to limit their colonization or pathogenic potential. This will include how innate and adaptive immune pathways target fungi and orchestrate antifungal immune responses, in addition to how secreted immune effectors, such as mucus and antimicrobial peptides, regulate fungal colonization and inhibit pathogenic potential. These immune interactions will be framed around our current understanding of the fungal effectors and pathways regulating colonization and pathogenesis within this niche. Finally, we highlight important unexplored mechanisms by which the immune system regulates commensal fungi in the gut.
Collapse
Affiliation(s)
- Owen Jensen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emma Trujillo
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Luke Hanson
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kyla S. Ost
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
2
|
Walsh D, Parmenter C, Bakker SE, Lithgow T, Traven A, Harrison F. A new model of endotracheal tube biofilm identifies combinations of matrix-degrading enzymes and antimicrobials able to eradicate biofilms of pathogens that cause ventilator-associated pneumonia. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001480. [PMID: 39088248 PMCID: PMC11541551 DOI: 10.1099/mic.0.001480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024]
Abstract
Ventilator-associated pneumonia is defined as pneumonia that develops in a patient who has been on mechanical ventilation for more than 48 hours through an endotracheal tube. It is caused by biofilm formation on the indwelling tube, which introduces pathogenic microbes such as Pseudomonas aeruginosa, Klebsiella pneumoniae and Candida albicans into the patient's lower airways. Currently, there is a lack of accurate in vitro models of ventilator-associated pneumonia development. This greatly limits our understanding of how the in-host environment alters pathogen physiology and the efficacy of ventilator-associated pneumonia prevention or treatment strategies. Here, we showcase a reproducible model that simulates the biofilm formation of these pathogens in a host-mimicking environment and demonstrate that the biofilm matrix produced differs from that observed in standard laboratory growth medium. In our model, pathogens are grown on endotracheal tube segments in the presence of a novel synthetic ventilated airway mucus medium that simulates the in-host environment. Matrix-degrading enzymes and cryo-scanning electron microscopy were employed to characterize the system in terms of biofilm matrix composition and structure, as compared to standard laboratory growth medium. As seen in patients, the biofilms of ventilator-associated pneumonia pathogens in our model either required very high concentrations of antimicrobials for eradication or could not be eradicated. However, combining matrix-degrading enzymes with antimicrobials greatly improved the biofilm eradication of all pathogens. Our in vitro endotracheal tube model informs on fundamental microbiology in the ventilator-associated pneumonia context and has broad applicability as a screening platform for antibiofilm measures including the use of matrix-degrading enzymes as antimicrobial adjuvants.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Chris Parmenter
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham, UK
| | | | - Trevor Lithgow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
- Center To Impact AMR, Monash University, Clayton 3800, Victoria, Australia
| | - Freya Harrison
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
3
|
Barbosa PF, Gonçalves DS, Ramos LS, Mello TP, Braga-Silva LA, Pinto MR, Taborda CP, Branquinha MH, Santos ALS. Saps1-3 Antigens in Candida albicans: Differential Modulation Following Exposure to Soluble Proteins, Mammalian Cells, and Infection in Mice. Infect Dis Rep 2024; 16:572-586. [PMID: 39051243 PMCID: PMC11270244 DOI: 10.3390/idr16040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
The secreted aspartic peptidases (Saps) of Candida albicans play crucial roles in various steps of fungal-host interactions. Using a flow cytometry approach, this study investigated the expression of Saps1-3 antigens after (i) incubation with soluble proteins, (ii) interaction with mammalian cells, and (iii) infection in immunosuppressed BALB/c mice. Supplementation strategies involving increasing concentrations of bovine serum albumin (BSA) added to yeast carbon base (YCB) medium as the sole nitrogenous source revealed a positive and significant correlation between BSA concentration and both the growth rate and the percentage of fluorescent cells (%FC) labeled with anti-Saps1-3 antibodies. Supplementing the YCB medium with various soluble proteins significantly modulated the expression of Saps1-3 antigens in C. albicans. Specifically, immunoglobulin G, gelatin, and total bovine/human sera significantly reduced the %FC, while laminin, human serum albumin, fibrinogen, hemoglobin, and mucin considerably increased the %FC compared to BSA. Furthermore, co-cultivating C. albicans yeasts with either live epithelial or macrophage cells induced the expression of Saps1-3 antigens in 78% (mean fluorescence intensity [MFI] = 152.1) and 82.7% (MFI = 178.2) of the yeast cells, respectively, compared to BSA, which resulted in 29.3% fluorescent cells (MFI = 50.9). Lastly, the yeasts recovered from the kidneys of infected immunosuppressed mice demonstrated a 4.8-fold increase in the production of Saps1-3 antigens (MFI = 246.6) compared to BSA, with 95.5% of yeasts labeled with anti-Saps1-3 antibodies. Altogether, these results demonstrated the positive modulation of Saps' expression in C. albicans by various key host proteinaceous components, as well as by in vitro and in vivo host challenges.
Collapse
Affiliation(s)
- Pedro F. Barbosa
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Diego S. Gonçalves
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Thaís P. Mello
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
| | - Lys A. Braga-Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Marcia R. Pinto
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense (UFF), Niterói 24210-130, Brazil;
| | - Carlos P. Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo 05508-060, Brazil;
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (P.F.B.); (D.S.G.); (L.S.R.); (T.P.M.); (L.A.B.-S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
- Rede Micologia RJ—Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro 21941-901, Brazil
| |
Collapse
|
4
|
Kaliniak S, Fiedoruk K, Spałek J, Piktel E, Durnaś B, Góźdź S, Bucki R, Okła S. Remodeling of Paranasal Sinuses Mucosa Functions in Response to Biofilm-Induced Inflammation. J Inflamm Res 2024; 17:1295-1323. [PMID: 38434581 PMCID: PMC10906676 DOI: 10.2147/jir.s443420] [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: 10/25/2023] [Accepted: 01/23/2024] [Indexed: 03/05/2024] Open
Abstract
Rhinosinusitis (RS) is an acute (ARS) or chronic (CRS) inflammatory disease of the nasal and paranasal sinus mucosa. CRS is a heterogeneous condition characterized by distinct inflammatory patterns (endotypes) and phenotypes associated with the presence (CRSwNP) or absence (CRSsNP) of nasal polyps. Mucosal barrier and mucociliary clearance dysfunction, inflammatory cell infiltration, mucus hypersecretion, and tissue remodeling are the hallmarks of CRS. However, the underlying factors, their priority, and the mechanisms of inflammatory responses remain unclear. Several hypotheses have been proposed that link CRS etiology and pathogenesis with host (eg, "immune barrier") and exogenous factors (eg, bacterial/fungal pathogens, dysbiotic microbiota/biofilms, or staphylococcal superantigens). The abnormal interplay between these factors is likely central to the pathophysiology of CRS by triggering compensatory immune responses. Here, we discuss the role of the sinonasal microbiota in CRS and its biofilms in the context of mucosal zinc (Zn) deficiency, serving as a possible unifying link between five host and "bacterial" hypotheses of CRS that lead to sinus mucosa remodeling. To date, no clear correlation between sinonasal microbiota and CRS has been established. However, the predominance of Corynebacteria and Staphylococci and their interspecies relationships likely play a vital role in the formation of the CRS-associated microbiota. Zn-mediated "nutritional immunity", exerted via calprotectin, alongside the dysregulation of Zn-dependent cellular processes, could be a crucial microbiota-shaping factor in CRS. Similar to cystic fibrosis (CF), the role of SPLUNC1-mediated regulation of mucus volume and pH in CRS has been considered. We complement the biofilms' "mechanistic" and "mucin" hypotheses behind CRS pathogenesis with the "structural" one - associated with bacterial "corncob" structures. Finally, microbiota restoration approaches for CRS prevention and treatment are reviewed, including pre- and probiotics, as well as Nasal Microbiota Transplantation (NMT).
Collapse
Affiliation(s)
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Jakub Spałek
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Bonita Durnaś
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Stanisław Góźdź
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| | - Sławomir Okła
- Holy-Cross Cancer Center, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, 25-317, Poland
| |
Collapse
|
5
|
Yang M, Xie M, Guo J, Zhang Y, Qiu Y, Wang Z, Du Y. Mucus-Permeable Sonodynamic Therapy Mediated Amphotericin B-Loaded PEGylated PLGA Nanoparticles Enable Eradication of Candida albicans Biofilm. Int J Nanomedicine 2023; 18:7941-7963. [PMID: 38169688 PMCID: PMC10758343 DOI: 10.2147/ijn.s437726] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Background Candida albicans (C. albicans) forms pathogenic biofilms, and the dense mucus layer secreted by the epithelium is a major barrier to the traditional antibiotic treatment of mucosa-associated C. albicans infections. Herein, we report a novel anti-biofilm strategy of mucus-permeable sonodynamic therapy (mp-SDT) based on ultrasound (US)-mediated amphotericin B-loaded PEGylated PLGA nanoparticles (AmB-NPs) to overcome mucus barrier and enable the eradication of C. albicans biofilm. Methods AmB-NPs were fabricated using ultrasonic double emulsion method, and their physicochemical and sonodynamic properties were determined. The mucus and biofilm permeability of US-mediated AmB-NPs were further investigated. Moreover, the anti-biofilm effect of US-mediated AmB-NPs treatment was thoroughly evaluated on mucus barrier abiotic biofilm, epithelium-associated biotic biofilm, and C. albicans-induced rabbit vaginal biofilms model. In addition, the ultrastructure and secreted cytokines of epithelial cells and the polarization of macrophages were analyzed to investigate the regulation of local cellular immune function by US-mediated AmB-NPs treatment. Results Polymeric AmB-NPs display excellent sonodynamic performance with massive singlet oxygen (1O2) generation. US-mediated AmB-NPs could rapidly transport through mucus and promote permeability in biofilms, which exhibited excellent eradicating ability to C. albicans biofilms. Furthermore, in the vaginal epithelial cells (VECs)-associated C. albicans biofilm model, the mp-SDT scheme showed the strongest biofilm eradication effect, with up to 98% biofilm re-formation inhibition rate, improved the ultrastructural damage, promoted local immune defense enhancement of VECs, and regulated the polarization of macrophages to the M1 phenotype to enhance macrophage-associated antifungal immune responses. In addition, mp-SDT treatment exhibited excellent therapeutic efficacy against C. albicans-induced rabbit vaginitis, promoted the recovery of mucosal epithelial ultrastructure, and contributed to the reshaping of a healthier vaginal microbiome. Conclusion The synergistic anti-biofilm strategies of mp-SDT effectively eradicated C. albicans biofilm and simultaneously regulated local antifungal immunity enhancement, which may provide a new approach to treat refractory drug-resistant biofilm-associated mucosal candidiasis.
Collapse
Affiliation(s)
- Min Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Mengyao Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jiajun Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuqing Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yan Qiu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
6
|
Hao Y, Wang T, Hou Y, Wang X, Yin Y, Liu Y, Han N, Ma Y, Li Z, Wei Y, Feng W, Jia Z, Qi H. Therapeutic potential of Lianhua Qingke in airway mucus hypersecretion of acute exacerbation of chronic obstructive pulmonary disease. Chin Med 2023; 18:145. [PMID: 37924136 PMCID: PMC10623880 DOI: 10.1186/s13020-023-00851-4] [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: 06/25/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Lianhua Qingke (LHQK) is an effective traditional Chinese medicine used for treating acute tracheobronchitis. In this study, we evaluated the effectiveness of LHQK in managing airway mucus hypersecretion in the acute exacerbation of chronic obstructive pulmonary disease (AECOPD). METHODS The AECOPD model was established by subjecting male Wistar rats to 12 weeks of cigarette smoke (CS) exposure (80 cigarettes/day, 5 days/week for 12 weeks) and intratracheal lipopolysaccharide (LPS) exposure (200 μg, on days 1, 14, and 84). The rats were divided into six groups: control (room air exposure), model (CS + LPS exposure), LHQK (LHQK-L, LHQK-M, and LHQK-H), and a positive control group (Ambroxol). H&E staining, and AB-PAS staining were used to evaluate lung tissue pathology, inflammatory responses, and goblet cell hyperplasia. RT-qPCR, immunohistochemistry, immunofluorescence and ELISA were utilized to analyze the transcription, expression and secretion of proteins related to mucus production in vivo and in the human airway epithelial cell line NCI-H292 in vitro. To predict and screen the active ingredients of LHQK, network pharmacology analysis and NF-κB reporter system analysis were employed. RESULTS LHQK treatment could ameliorate AECOPD-triggered pulmonary structure damage, inflammatory cell infiltration, and pro-inflammatory cytokine production. AB-PAS and immunofluorescence staining with CCSP and Muc5ac antibodies showed that LHQK reduced goblet cell hyperplasia, probably by inhibiting the transdifferentiation of Club cells into goblet cells. RT-qPCR and immunohistochemistry of Muc5ac and APQ5 showed that LHQK modulated mucus homeostasis by suppressing Muc5ac transcription and hypersecretion in vivo and in vitro, and maintaining the balance between Muc5ac and AQP5 expression. Network pharmacology analysis and NF-κB luciferase reporter system analysis provided insights into the active ingredients of LHQK that may help control airway mucus hypersecretion and regulate inflammation. CONCLUSION LHQK demonstrated therapeutic effects in AECOPD by reducing inflammation, suppressing goblet cell hyperplasia, preventing Club cell transdifferentiation, reducing Muc5ac hypersecretion, and modulating airway mucus homeostasis. These findings support the clinical use of LHQK as a potential treatment for AECOPD.
Collapse
Affiliation(s)
- Yuanjie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Yunlong Hou
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Xiaoqi Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Yujie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yan Ma
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yaru Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuan, 050090, Hebei, China
| | - Wei Feng
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Zhenhua Jia
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
- Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, 050091, Hebei, China.
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, Hebei, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| |
Collapse
|
7
|
Kaur J, Nobile CJ. Antifungal drug-resistance mechanisms in Candida biofilms. Curr Opin Microbiol 2023; 71:102237. [PMID: 36436326 PMCID: PMC11569868 DOI: 10.1016/j.mib.2022.102237] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 11/27/2022]
Abstract
Infections caused by the Candida species of human fungal pathogens are a significant medical problem because they can disseminate to nearly every organ of the body. In addition, there are only a few classes of antifungal drugs available to treat patients with invasive fungal infections. Candida infections that are associated with biofilms can withstand much higher concentrations of antifungal drugs compared with infections caused by planktonic cells, thus making biofilm infections particularly challenging to treat. Candida albicans is among the most prevalent fungal species of the human microbiota, asymptomatically colonizing several niches of the body, including the gastrointestinal tract, genitourinary tract, mouth, and skin. Immunocompromised health conditions, dysbiosis of the microbiota, or environmental changes, however, can lead to C. albicans overgrowth, causing infections that range from superficial mucosal infections to severe hematogenously disseminated infections. Here, we review the current knowledge of antifungal drug-resistance mechanisms occurring in Candida biofilms.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA; Health Sciences Research Institute, University of California Merced, Merced, CA, USA.
| |
Collapse
|
8
|
Bi Q, Song X, Zhao Y, Hu X, Yang H, Jin R, Nie Y. Mucus-penetrating nonviral gene vaccine processed in the epithelium for inducing advanced vaginal mucosal immune responses. Acta Pharm Sin B 2022; 13:1287-1302. [PMID: 36970203 PMCID: PMC10031263 DOI: 10.1016/j.apsb.2022.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/29/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022] Open
Abstract
Establishment of vaginal immune defenses at the mucosal interface layer through gene vaccines promise to prevent infectious diseases among females. Mucosal barriers composed of a flowing mucus hydrogel and tightly conjugated epithelial cells (ECs), which represent the main technical difficulties for vaccine development, reside in the harsh, acidic human vaginal environment. Different from frequently employed viral vectors, two types of nonviral nanocarriers were designed to concurrently overcome the barriers and induce immune responses. Differing design concepts include the charge-reversal property (DRLS) to mimic a virus that uses any cells as factories, as well as the addition of a hyaluronic acid coating (HA/RLS) to directly target dendritic cells (DCs). With a suitable size and electrostatic neutrality, these two nanoparticles penetrate a mucus hydrogel with similar diffusivity. The DRLS system expressed a higher level of the carried human papillomavirus type 16 L1 gene compared to HA/RLS in vivo. Therefore it induced more robust mucosal, cellular, and humoral immune responses. Moreover, the DLRS applied to intravaginal immunization induced high IgA levels compared with intramuscularly injected DNA (naked), indicating timely protection against pathogens at the mucus layer. These findings also offer important approaches for the design and fabrication of nonviral gene vaccines in other mucosal systems.
Collapse
|
9
|
Łysik D, Deptuła P, Chmielewska S, Bucki R, Mystkowska J. Degradation of Polylactide and Polycaprolactone as a Result of Biofilm Formation Assessed under Experimental Conditions Simulating the Oral Cavity Environment. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7061. [PMID: 36295125 PMCID: PMC9604997 DOI: 10.3390/ma15207061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023]
Abstract
Polylactide (PLA) and polycaprolactone (PCL) are biodegradable and bioabsorbable thermoplastic polymers considered as promising materials for oral applications. However, any abiotic surface used, especially in areas naturally colonized by microorganisms, provides a favorable interface for microbial growth and biofilm development. In this study, we investigated the biofilm formation of C. krusei and S. mutans on the surface of PLA and PCL immersed in the artificial saliva. Using microscopic (AFM, CLSM) observations and spectrometric measurements, we assessed the mass and topography of biofilm that developed on PLA and PCL surfaces. Incubated up to 56 days in specially prepared saliva and microorganisms medium, solid polymer samples were examined for surface properties (wettability, roughness, elastic modulus of the surface layer), structure (molecular weight, crystallinity), and mechanical properties (hardness, tensile strength). It has been shown that biofilm, especially S. mutans, promotes polymer degradation. Our findings indicate the need for additional antimicrobial strategies for the effective oral applications of PLA and PCL.
Collapse
Affiliation(s)
- Dawid Łysik
- Institute of Biomedical Engineering, Bialystok University of Technology, 15-351 Bialystok, Poland
| | - Piotr Deptuła
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Sylwia Chmielewska
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Joanna Mystkowska
- Institute of Biomedical Engineering, Bialystok University of Technology, 15-351 Bialystok, Poland
| |
Collapse
|
10
|
Takagi J, Aoki K, Turner BS, Lamont S, Lehoux S, Kavanaugh N, Gulati M, Valle Arevalo A, Lawrence TJ, Kim CY, Bakshi B, Ishihara M, Nobile CJ, Cummings RD, Wozniak DJ, Tiemeyer M, Hevey R, Ribbeck K. Mucin O-glycans are natural inhibitors of Candida albicans pathogenicity. Nat Chem Biol 2022; 18:762-773. [PMID: 35668191 PMCID: PMC7613833 DOI: 10.1038/s41589-022-01035-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Mucins are large gel-forming polymers inside the mucus barrier that inhibit the yeast-to-hyphal transition of Candida albicans, a key virulence trait of this important human fungal pathogen. However, the molecular motifs in mucins that inhibit filamentation remain unclear despite their potential for therapeutic interventions. Here, we determined that mucins display an abundance of virulence-attenuating molecules in the form of mucin O-glycans. We isolated and cataloged >100 mucin O-glycans from three major mucosal surfaces and established that they suppress filamentation and related phenotypes relevant to infection, including surface adhesion, biofilm formation and cross-kingdom competition between C. albicans and the bacterium Pseudomonas aeruginosa. Using synthetic O-glycans, we identified three structures (core 1, core 1 + fucose and core 2 + galactose) that are sufficient to inhibit filamentation with potency comparable to the complex O-glycan pool. Overall, this work identifies mucin O-glycans as host molecules with untapped therapeutic potential to manage fungal pathogens.
Collapse
Affiliation(s)
- Julie Takagi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Bradley S Turner
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sabrina Lamont
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH, USA
| | - Sylvain Lehoux
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, National Center for Functional Glycomics, Boston, MA, USA
| | - Nicole Kavanaugh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Megha Gulati
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Molecular Cell, Cell Press, Cambridge, MA, USA
| | - Ashley Valle Arevalo
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, CA, USA
| | - Travis J Lawrence
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, CA, USA
- Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Colin Y Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bhavya Bakshi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
- Health Sciences Research Institute, University of California Merced, Merced, CA, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, National Center for Functional Glycomics, Boston, MA, USA
| | - Daniel J Wozniak
- Departments of Microbial Infection and Immunity, Microbiology, The Ohio State University, Columbus, OH, USA
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Rachel Hevey
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| | - Katharina Ribbeck
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
11
|
Wu X, Gu B, Yang H. The role of γδ T cells in the interaction between commensal and pathogenic bacteria in the intestinal mucosa. Int Rev Immunol 2022; 42:379-392. [PMID: 35583374 DOI: 10.1080/08830185.2022.2076846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 12/22/2022]
Abstract
The intestinal mucosa is an important structure involved in resistance to pathogen infection. It is mainly composed of four barriers, which have different but interrelated functions. Pathogenic bacteria can damage these intestinal mucosal barriers. Here, we mainly review the mechanisms of pathogen damage to biological barriers. Most γδ T cells are located on the surface of the intestinal mucosa, with the ability to migrate and engage in crosstalk with microorganisms. Commensal bacteria are involved in the activation and migration of γδ T cells to monitor the invasion of pathogens. Pathogen invasion alters the migration pattern of γδ T cells. γδ T cells accelerate pathogen clearance and limit opportunistic invasion of commensal bacteria. By discussing these interactions among γδ T cells, commensal bacteria and pathogenic bacteria, we suggest that γδ T cells may link the interactions between commensal bacteria and pathogenic bacteria.
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bing Gu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
12
|
González-Orozco BD, García-Cano I, Jiménez-Flores R, Alvárez VB. Invited review: Milk kefir microbiota—Direct and indirect antimicrobial effects. J Dairy Sci 2022; 105:3703-3715. [DOI: 10.3168/jds.2021-21382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/10/2022] [Indexed: 11/19/2022]
|
13
|
Maciel EI, Valle Arevalo A, Ziman B, Nobile CJ, Oviedo NJ. Epithelial Infection With Candida albicans Elicits a Multi-System Response in Planarians. Front Microbiol 2021; 11:629526. [PMID: 33519792 PMCID: PMC7840899 DOI: 10.3389/fmicb.2020.629526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
Candida albicans is one of the most common fungal pathogens of humans. Prior work introduced the planarian Schmidtea mediterranea as a new model system to study the host response to fungal infection at the organismal level. In the current study, we analyzed host-pathogen changes that occurred in situ during early infection with C. albicans. We found that the transcription factor Bcr1 and its downstream adhesin Als3 are required for C. albicans to adhere to and colonize the planarian epithelial surface, and that adherence of C. albicans triggers a multi-system host response that is mediated by the Dectin signaling pathway. This infection response is characterized by two peaks of stem cell divisions and transcriptional changes in differentiated tissues including the nervous and the excretory systems. This response bears some resemblance to a wound-like response to physical injury; however, it takes place without visible tissue damage and it engages a distinct set of progenitor cells. Overall, we identified two C. albicans proteins that mediate epithelial infection of planarians and a comprehensive host response facilitated by diverse tissues to effectively clear the infection.
Collapse
Affiliation(s)
- Eli Isael Maciel
- Department of Molecular & Cell Biology, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Ashley Valle Arevalo
- Department of Molecular & Cell Biology, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Benjamin Ziman
- Department of Molecular & Cell Biology, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Clarissa J Nobile
- Department of Molecular & Cell Biology, University of California, Merced, Merced, CA, United States.,Health Sciences Research Institute, University of California, Merced, Merced, CA, United States
| | - Néstor J Oviedo
- Department of Molecular & Cell Biology, University of California, Merced, Merced, CA, United States.,Health Sciences Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
14
|
Černáková L, Rodrigues CF. Microbial interactions and immunity response in oral Candida species. Future Microbiol 2020; 15:1653-1677. [PMID: 33251818 DOI: 10.2217/fmb-2020-0113] [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] [Indexed: 12/12/2022] Open
Abstract
Oral candidiasis are among the most common noncommunicable diseases, related with serious local and systemic illnesses. Although these infections can occur in all kinds of patients, they are more recurrent in immunosuppressed ones such as patients with HIV, hepatitis, cancer or under long antimicrobial treatments. Candida albicans continues to be the most frequently identified Candida spp. in these disorders, but other non-C. albicans Candida are rising. Understanding the immune responses involved in oral Candida spp. infections is a key feature to a successful treatment and to the design of novel therapies. In this review, we performed a literature search in PubMed and WoS, in order to examine and analyze common oral Candida spp.-bacteria/Candida-Candida interactions and the host immunity response in oral candidiasis.
Collapse
Affiliation(s)
- Lucia Černáková
- Department of Microbiology & Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Célia F Rodrigues
- Department of Chemical Engineering, LEPABE - Laboratory for Process Engineering, Environment, Biotechnology & Energy, Faculty of Engineering, University of Porto, Portugal
| |
Collapse
|