1
|
Shahdab N, Ward C, Hansbro PM, Cummings S, Young JS, Moheimani F. Distinct Effects of Respiratory Viral Infection Models on miR-149-5p, IL-6 and p63 Expression in BEAS-2B and A549 Epithelial Cells. Cells 2024; 13:919. [PMID: 38891051 PMCID: PMC11172188 DOI: 10.3390/cells13110919] [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: 04/27/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Respiratory viruses cause airway inflammation, resulting in epithelial injury and repair. miRNAs, including miR-149-5p, regulate different pathological conditions. We aimed to determine how miR-149-5p functions in regulating pro-inflammatory IL-6 and p63, key regulators of airway epithelial wound repair, in response to viral proteins in bronchial (BEAS-2B) and alveolar (A549) epithelial cells. BEAS-2B or A549 cells were incubated with poly (I:C, 0.5 µg/mL) for 48 h or SARS-CoV-2 spike protein-1 or 2 subunit (S1 or S2, 1 μg/mL) for 24 h. miR-149-5p was suppressed in BEAS-2B challenged with poly (I:C), correlating with IL-6 and p63 upregulation. miR-149-5p was down-regulated in A549 stimulated with poly (I:C); IL-6 expression increased, but p63 protein levels were undetectable. miR-149-5p remained unchanged in cells exposed to S1 or S2, while S1 transfection increased IL-6 expression in BEAS-2B cells. Ectopic over-expression of miR-149-5p in BEAS-2B cells suppressed IL-6 and p63 mRNA levels and inhibited poly (I:C)-induced IL-6 and p63 mRNA expressions. miR-149-5p directly suppressed IL-6 mRNA in BEAS-2B cells. Hence, BEAS-2B cells respond differently to poly (I:C), S1 or S2 compared to A549 cells. Thus, miR-149-5p dysregulation may be involved in poly (I:C)-stimulated but not S1- or S2-stimulated increased IL-6 production and p63 expression in BEAS-2B cells.
Collapse
Affiliation(s)
- Nafeesa Shahdab
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Christopher Ward
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia;
| | - Stephen Cummings
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - John S. Young
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Fatemeh Moheimani
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
| |
Collapse
|
2
|
Keulers L, Dehghani A, Knippels L, Garssen J, Papadopoulos N, Folkerts G, Braber S, van Bergenhenegouwen J. Probiotics, prebiotics, and synbiotics to prevent or combat air pollution consequences: The gut-lung axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119066. [PMID: 35240267 DOI: 10.1016/j.envpol.2022.119066] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 05/26/2023]
Abstract
Air pollution exposure is a public health emergency, which attributes globally to an estimated seven million deaths on a yearly basis We are all exposed to air pollutants, varying from ambient air pollution hanging over cities to dust inside the home. It is a mixture of airborne particulate matter and gases that can be subdivided into three categories based on particle diameter. The smallest category called PM0.1 is the most abundant. A fraction of the particles included in this category might enter the blood stream spreading to other parts of the body. As air pollutants can enter the body via the lungs and gut, growing evidence links its exposure to gastrointestinal and respiratory impairments and diseases, like asthma, rhinitis, respiratory tract infections, Crohn's disease, ulcerative colitis, and abdominal pain. It has become evident that there exists a crosstalk between the respiratory and gastrointestinal tracts, commonly referred to as the gut-lung axis. Via microbial secretions, metabolites, immune mediators and lipid profiles, these two separate organ systems can influence each other. Well-known immunomodulators and gut health stimulators are probiotics, prebiotics, together called synbiotics. They might combat air pollution-induced systemic inflammation and oxidative stress by optimizing the microbiota composition and microbial metabolites, thereby stimulating anti-inflammatory pathways and strengthening mucosal and epithelial barriers. Although clinical studies investigating the role of probiotics, prebiotics, and synbiotics in an air pollution setting are lacking, these interventions show promising health promoting effects by affecting the gastrointestinal- and respiratory tract. This review summarizes the current data on how air pollution can affect the gut-lung axis and might impact gut and lung health. It will further elaborate on the potential role of probiotics, prebiotics and synbiotics on the gut-lung axis, and gut and lung health.
Collapse
Affiliation(s)
- Loret Keulers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands.
| | - Ali Dehghani
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Leon Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Nikolaos Papadopoulos
- Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Oxford Road M13 9PL, Manchester, United Kingdom
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| |
Collapse
|
3
|
Gweon B, Jang TK, Thuy PX, Moon EY. Primary Cilium by Polyinosinic:Polycytidylic Acid Regulates the Regenerative Migration of Beas-2B Bronchial Epithelial Cells. Biomol Ther (Seoul) 2022; 30:170-178. [PMID: 35221299 PMCID: PMC8902458 DOI: 10.4062/biomolther.2022.009] [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: 01/22/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 11/17/2022] Open
Abstract
The airway epithelium is equipped with the ability to resist respiratory disease development and airway damage, including the migration of airway epithelial cells and the activation of TLR3, which recognizes double-stranded (ds) RNA. Primary cilia on airway epithelial cells are involved in the cell cycle and cell differentiation and repair. In this study, we used Beas-2B human bronchial epithelial cells to investigate the effects of the TLR3 agonist polyinosinic:polycytidylic acid [Poly(I:C)] on airway cell migration and primary cilia (PC) formation. PC formation increased in cells incubated under serum deprivation. Migration was faster in Beas-2B cells pretreated with Poly(I:C) than in control cells, as judged by a wound healing assay, single-cell path tracking, and a Transwell migration assay. No changes in cell migration were observed when the cells were incubated in conditioned medium from Poly(I:C)-treated cells. PC formation was enhanced by Poly(I:C) treatment, but was reduced when the cells were exposed to the ciliogenesis inhibitor ciliobrevin A (CilioA). The inhibition of Beas-2B cell migration by CilioA was also assessed and a slight decrease in ciliogenesis was detected in SARS-CoV-2 spike protein (SP)-treated Beas-2B cells overexpressing ACE2 compared to control cells. Cell migration was decreased by SP but restored by Poly(I:C) treatment. Taken together, our results demonstrate that impaired migration by SP-treated cells can be attenuated by Poly(I:C) treatment, thus increasing airway cell migration through the regulation of ciliogenesis.
Collapse
Affiliation(s)
- Bomi Gweon
- Department of Mechanical Engineering, Sejong University, Seoul 05006, Republic of Korea
| | - Tae-Kyu Jang
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Pham Xuan Thuy
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Eun-Yi Moon
- Department of Integrated Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
4
|
Lan X, Zhang W, Zhu J, Huang H, Mo K, Guo H, Zhu L, Liu J, Li M, Wang L, Liu C, Ji J, Ouyang H. dsRNA Induced IFNβ-MMP13 Axis Drives Corneal Wound Healing. Invest Ophthalmol Vis Sci 2022; 63:14. [PMID: 35129588 PMCID: PMC8822365 DOI: 10.1167/iovs.63.2.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Cornea, the outermost transparent layer of the eye, is the first line of defense against external threats. Following injury, the wound healing response is crucial to corneal repair and regeneration, yet its underlying mechanism is poorly understood. Our study was designed to investigate the role of dsRNA and its regulatory network in corneal wound healing. Methods A corneal wound healing model was established via the surgical removal of half of the corneal surface and adjoining limbus. RNase III was then used to clarify the role of dsRNA in corneal wound closure and RNA-seq was performed to investigate the mechanism of dsRNA in the healing process. Related gene expression was assessed using immunofluorescence staining, qPCR, and Western blot. Flow cytometry and scratch assay were used to analyze the proliferation and migration of limbal stem/progenitor cells (LSCs) in vitro and functional analysis of the target genes was completed using the corneal wound healing model. Results Corneal wound healing was delayed and impaired when the dsRNAs were removed or damaged following RNase III digestion. The dsRNAs released following corneal damage activate type I interferon (IFN-I) signaling, primarily IFNβ, via the corneal epithelium and neutralizing IFNβ or blocking IFN-I signaling delays corneal wound closure. Moreover, our data identified MMP13 as a downstream effector of IFNβ where its expression promotes LSC proliferation and enhances corneal epithelial reconstruction in vivo. Conclusions The dsRNA induced IFNβ-MMP13 axis plays a key role in corneal wound healing.
Collapse
Affiliation(s)
- Xihong Lan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wang Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jin Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huaxing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Kunlun Mo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huizhen Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Liqiong Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiafeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Mingsen Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chunqiao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianping Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Hong Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Silveira MLC, Tamashiro E, Santos ARD, Martins RB, Faria FM, Silva LECM, Torrieri R, de C Ruy P, Silva WA, Arruda E, Anselmo-Lima WT, Valera FCP. miRNA-205-5p can be related to T2-polarity in Chronic Rhinosinusitis with Nasal Polyps. Rhinology 2021; 59:567-576. [PMID: 34608897 DOI: 10.4193/rhin21.109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND microRNAs (miRNAs) are directly associated with inflammatory response, but their direct role in CRSwNP (chronic rhinosinusitis with nasal polyps) remains evasive. This study aimed to compare the expression of several miRNAs in tissue samples obtained from patients with CRSwNP and controls and to evaluate if miRNAs correlate to a specific inflammatory pattern (T1, T2, T17, and Treg) or intensity of symptoms in CRSwNP. METHODS nasal polyps (from patients with CRSwNP - n=36) and middle turbinate mucosa (from control patients - n=41) were collected. Microarray determined human mature miRNA expression, and the results obtained were validated by qPCR. miRNAs that were differentially expressed were then correlated to cytokine proteins (by Luminex), tissue eosinophilia, and SNOT-22. RESULTS After microarray and qPCR analyses, six microRNAs were up-regulated in CRSwNP samples when compared with controls: miR-205-5p, miR-221-3p, miR-222-3p, miR-378a-3p, miR-449a and miR-449b-5p. All these miRNAs are directly implicated with cell cycle regulation and apoptosis, and to a minor extent, with inflammation. Importantly, miR-205-5p showed a significantly positive correlation with IL-5 concentration and eosinophil count at the tissue and with the worst SNOT-22 score. CONCLUSIONS miRNA 205-5p was increased in CRSwNP compared to controls, and it was especially expressed in CRSwNP patients with higher T2 inflammation (measured by both IL-5 levels and local eosinophilia) and worst clinical presentation. This miRNA may be an interesting target to be explored in patients with CRSwNP.
Collapse
Affiliation(s)
- M L C Silveira
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - E Tamashiro
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - A R D Santos
- Genomics Medical Center, Clinics Hospital at Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - R B Martins
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - F M Faria
- Department of Pathology and Legal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - L E C M Silva
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - R Torrieri
- Genomics Medical Center, Clinics Hospital at Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - P de C Ruy
- Genomics Medical Center, Clinics Hospital at Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - W A Silva
- Genomics Medical Center, Clinics Hospital at Ribeirao Preto Medical School, University of Sao Paulo, Brazil.,Department of Genetics, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - E Arruda
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Virology Research Center, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - W T Anselmo-Lima
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - F C P Valera
- Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| |
Collapse
|
6
|
Chen Q, Tan KS, Liu J, Ong HH, Zhou S, Huang H, Chen H, Ong YK, Thong M, Chow VT, Qiu Q, Wang DY. Host Antiviral Response Suppresses Ciliogenesis and Motile Ciliary Functions in the Nasal Epithelium. Front Cell Dev Biol 2020; 8:581340. [PMID: 33409274 PMCID: PMC7779769 DOI: 10.3389/fcell.2020.581340] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
Background Respiratory viral infections are one of the main drivers of development and exacerbation for chronic airway inflammatory diseases. Increased viral susceptibility and impaired mucociliary clearance are often associated with chronic airway inflammatory diseases and served as risk factors of exacerbations. However, the links between viral susceptibility, viral clearance, and impaired mucociliary functions are unclear. Therefore, the objective of this study is to provide the insights into the effects of improper clearance of respiratory viruses from the epithelium following infection, and their resulting persistent activation of antiviral response, on mucociliary functions. Methods In order to investigate the effects of persistent antiviral responses triggered by viral components from improper clearance on cilia formation and function, we established an in vitro air–liquid interface (ALI) culture of human nasal epithelial cells (hNECs) and used Poly(I:C) as a surrogate of viral components to simulate their effects toward re-epithelization and mucociliary functions of the nasal epithelium following damages from a viral infection. Results Through previous and current viral infection expression data, we found that respiratory viral infection of hNECs downregulated motile cilia gene expression. We then further tested the effects of antiviral response activation on the differentiation of hNECs using Poly(I:C) stimulation on differentiating human nasal epithelial stem/progenitor cells (hNESPCs). Using this model, we observed reduced ciliated cell differentiation compared to goblet cells, reduced protein and mRNA in ciliogenesis-associated markers, and increased mis-assembly and mis-localization of ciliary protein DNAH5 following treatment with 25 μg/ml Poly(I:C) in differentiating hNECs. Additionally, the cilia length and ciliary beat frequency (CBF) were also decreased, which suggest impairment of ciliary function as well. Conclusion Our results suggest that the impairments of ciliogenesis and ciliary function in hNECs may be triggered by specific expression of host antiviral response genes during re-epithelization of the nasal epithelium following viral infection. This event may in turn drive the development and exacerbation of chronic airway inflammatory diseases.
Collapse
Affiliation(s)
- Qianmin Chen
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hsiao Hui Ong
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suizi Zhou
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hongming Huang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Otolaryngology, Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China
| | - Hailing Chen
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yew Kwang Ong
- Department of Otolaryngology, Head and Neck Surgery, National University Health System, National University Hospital, Singapore, Singapore
| | - Mark Thong
- Department of Otolaryngology, Head and Neck Surgery, National University Health System, National University Hospital, Singapore, Singapore
| | - Vincent T Chow
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Qianhui Qiu
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
7
|
Li L, Pan J, Cai X, Gong E, Xu C, Zheng H, Cao Z, Yin Z. Human umbilical cord mesenchymal stem cells suppress lung cancer via TLR4/NF-κB signalling pathway. BIOTECHNOL BIOTEC EQ 2020. [DOI: 10.1080/13102818.2020.1712257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Lu Li
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Jiongwei Pan
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Xiaoping Cai
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Enhui Gong
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Cunlai Xu
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Hao Zheng
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Zhuo Cao
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| | - Zhangyong Yin
- Respiratory Department, Lishui People’s Hospital, Lishui, P.R. China
| |
Collapse
|
8
|
Therapeutic effectiveness of type I interferon in vulvovaginal candidiasis. Microb Pathog 2019; 134:103562. [PMID: 31158491 DOI: 10.1016/j.micpath.2019.103562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/15/2019] [Accepted: 05/30/2019] [Indexed: 11/20/2022]
Abstract
Vulvovaginal candidiasis (VVC) affects approximately 75% of all women of during their reproductive years. Previously, we reported that recombinant human IFN α-2b (rhIFNα-2b) protects vaginal epithelial cells from candidal injury in vitro. In the current study, we examined the effects of rhIFNα-2b (1.25 mg/mL, 10% inhibition concentration) on fungal clearance, immunocompetent cytokine responses, non-B IgG production, and tissue repair in a rat model of VVC. Following rhIFNα-2b treatment, the negative pathogen conversion rate reached 50.0% (3/6). Although rhIFNα-2b exhibited a limited ability to decrease inflammation and injury progression (P > 0.05), the Flameng mitochondrial injury scores were significantly reduced (P < 0.001) compared with those of the Model rats. After rhIFNα-2b treatment, the levels of IFN-γ and epithelial-derived IgG (tested by RP215) in vaginal tissues were significantly increased with those in the Control and Model groups (both P < 0.001), while there were no significant differences in the levels of IL-4 and IL-17 (P > 0.05). This is the first study to address the efficacy of rhIFNα-2b in treating VVC in a rat model, providing a theoretical basis for development of this promising treatment for clinical use.
Collapse
|
9
|
Cowin AJ. New Innovations in Wound Healing and Repair. Int J Mol Sci 2019; 20:ijms20071724. [PMID: 30965550 PMCID: PMC6479748 DOI: 10.3390/ijms20071724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Allison J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide SA 5095, Australia.
| |
Collapse
|