1
|
Trivedi S, Deering-Rice CE, Aamodt SE, Huecksteadt TP, Myers EJ, Sanders KA, Paine R, Warren KJ. Progesterone amplifies allergic inflammation and airway pathology in association with higher lung ILC2 responses. Am J Physiol Lung Cell Mol Physiol 2024; 327:L65-L78. [PMID: 38651968 PMCID: PMC11380947 DOI: 10.1152/ajplung.00207.2023] [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: 07/07/2023] [Revised: 03/02/2024] [Accepted: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
Perimenstrual worsening of asthma occurs in up to 40% of women with asthma, leading to increased acute exacerbations requiring clinical care. The role of sex hormones during these times remains unclear. In the current study, we used a translational approach to determine whether progesterone exacerbates allergic inflammation in the traditional chicken egg ovalbumin (OVA) model in BALB/c mice. Simultaneously, we used peripheral blood mononuclear cells (PBMC) from healthy human donors to assess the effects of progesterone on circulating group 2 innate lymphoid cells (ILC2). Briefly, lungs of ovariectomized (OVX) or sham-operated female (F-Sham) controls were implanted with a progesterone (P4, 25 mg) (OVX-P4) or placebo pellet (OVX-Placebo), followed by sensitization and challenge with ovalbumin (OVA). Progesterone increased total inflammatory histologic scores, increased hyper-responsiveness to methacholine (MCh), increased select chemokines in the bronchoalveolar lavage (BAL) and serum, and increased ILC2 and neutrophil numbers, along the airways compared with F-Sham-OVA and OVX-Placebo-OVA animals. Lung ILC2 were sorted from F-Sham-OVA, OVX-Placebo-OVA and OVX-P4-OVA treated animals and stimulated with IL-33. OVX-P4-OVA lung ILC2 were more responsive to interleukin 33 (IL-33) compared with F-Sham-OVA treated, producing more IL-13 and chemokines following IL-33 stimulation. We confirmed the expression of the progesterone receptor (PR) on human ILC2, and showed that P4 + IL-33 stimulation also increased IL-13 and chemokine production from human ILC2. We establish that murine ILC2 are capable of responding to P4 and thereby contribute to allergic inflammation in the lung. We confirmed that human ILC2 are also hyper-responsive to P4 and IL-33 and likely contribute to airway exacerbations following allergen exposures in asthmatic women with increased symptoms around the time of menstruation.NEW & NOTEWORTHY There is a strong association between female biological sex and severe asthma. We investigated the allergic immune response, lung pathology, and airway mechanics in the well-described chicken egg ovalbumin (OVA) model with steady levels of progesterone delivered throughout the treatment period. We found that progesterone enhances the activation of mouse group 2 innate lymphoid cells (ILC2). Human ILC2 are also hyper-responsive to progesterone and interleukin 33 (IL-33), and likely contribute to airway exacerbations following allergen exposures in women with asthma.
Collapse
Affiliation(s)
- Shubhanshi Trivedi
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Samuel E Aamodt
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
| | - Thomas P Huecksteadt
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Elizabeth J Myers
- Division of Neuroimmunology, Department of Neurology, University of Utah Health, Salt Lake City, Utah, United States
| | - Karl A Sanders
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Robert Paine
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| | - Kristi J Warren
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, Salt Lake City, Utah, United States
- George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, United States
| |
Collapse
|
2
|
Li L, Zhang Y, Liu H, Wang T, Li J, Wang X. Exploring causal relationships between inflammatory cytokines and allergic rhinitis, chronic rhinosinusitis, and nasal polyps: a Mendelian randomization study. Front Immunol 2023; 14:1288517. [PMID: 38022554 PMCID: PMC10667686 DOI: 10.3389/fimmu.2023.1288517] [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: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Previous research has suggested connections between specific inflammatory cytokines and nasal conditions, including Allergic Rhinitis (AR), Chronic Rhinosinusitis (CRS), and Nasal Polyps (NP). However, a lack of robust research establishing the causal underpinnings of them. This Mendelian Randomization (MR) study aims to evaluate the causal relationships between 41 inflammatory cytokines and the incidence of AR, CRS and NP. Methods This study employed a two-sample MR design, harnessing genetic variations derived from publicly accessible genome-wide association studies (GWAS) datasets. AR data was sourced from a GWAS with 25,486 cases and 87,097 controls (identifier: ukb-b-7178). CRS data originated from a GWAS encompassing 1,179 cases and 360,015 controls (identifier: ukb-d-J32). NP data was extracted from a GWAS involving 1,637 cases and 335,562 controls (identifier: ukb-a-541). The data for 41 inflammatory cytokines were obtained from an independent GWAS encompassing 8,293 participants. Inverse variance weighted (IVW), MR Egger regression and Weighted median were used to evaluate the causalities of exposures and outcomes. A range of sensitivity analyses were implemented to assess the robustness of the results. Results The results revealed significant associations between elevated circulating levels of MIP-1α (odds ratio, OR: 1.01798, 95% confidence interval, CI: 1.00217-1.03404, p = 0.02570) and TNF-α (OR: 1.01478, 95% CI: 1.00225-1.02746, p = 0.02067) with an augmented risk of AR in the IVW approach. Heightened levels of circulating IL-2 exhibited a positive correlation with an increased susceptibility to NP in the IVW approach (OR: 1.00129, 95% CI: 1.00017-1.00242, p = 0.02434), whereas elevated levels of circulating PDGF-BB demonstrated a decreased risk of NP (OR: 0.99920, 95% CI: 0.99841-0.99999, p = 0.047610). The MR analysis between levels of 41 inflammatory cytokines and the incidence of CRS yielded no positive outcomes. Conclusion This investigation proposes a potential causal association between elevated levels of MIP-1α and TNF-α with an elevated risk of AR, as well as an increased risk of NP linked to elevated IL-2 levels. Furthermore, there appears to be a potential association between increased levels of circulating PDGF-BB and a reduced risk of NP.
Collapse
Affiliation(s)
- Li Li
- Department of Otolaryngology-Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Yuanding Zhang
- Department of Otolaryngology-Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Hong Liu
- Department of Otolaryngology-Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Tianqi Wang
- Department of Otolaryngology-Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Junxin Li
- Department of Rehabilitation Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xin Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Abstract
Systemic sclerosis (SSc) is a chronic immune-mediated disease characterized by microangiopathy, immune dysregulation, and progressive fibrosis of the skin and internal organs. Though not fully understood, the pathogenesis of SSc is dominated by microvascular injury, endothelial dysregulation, and immune response that are thought to be associated with fibroblast activation and related fibrogenesis. Among the main clinical subsets, diffuse SSc (dSSc) is a progressive form with rapid and disseminated skin thickening accompanied by internal organ fibrosis and dysfunction. Despite recent advances and multiple randomized clinical trials in early dSSc patients, an effective disease-modifying treatment for progressive skin fibrosis is still missing, and there is a crucial need to identify new targets for therapeutic intervention. Eotaxin-2 (CCL24) is a chemokine secreted by immune cells and epithelial cells, which promotes trafficking of immune cells and activation of pro-fibrotic cells through CCR3 receptor binding. Higher levels of CCL24 and CCR3 were found in the skin and sera of patients with SSc compared with healthy controls; elevated levels of CCL24 and CCR3 were associated with fibrosis and predictive of greater lung function deterioration. Growing evidence supports the potency of a CCL24-blocking antibody as an anti-inflammatory and anti-fibrotic modulating agent in multiple preclinical models that involve liver, skin, and lung inflammation and fibrosis. This review highlights the role of CCL24 in orchestrating immune, vascular, and fibrotic pathways, and the potential of CCL24 inhibition as a novel treatment for SSc.
Collapse
Affiliation(s)
| | | | - Alexandra Balbir-Gurman
- Rheumatology Institute, Rambam Health Care Campus, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
4
|
Farmanzadeh A, Qujeq D, Yousefi T. The Interaction Network of MicroRNAs with Cytokines and Signaling Pathways in Allergic Asthma. Microrna 2022; 11:104-117. [PMID: 35507792 DOI: 10.2174/2211536611666220428134324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/15/2022] [Accepted: 03/10/2022] [Indexed: 01/01/2023]
Abstract
Allergic asthma is a complicated disease that is affected by many factors. Numerous cytokines and signaling pathways are attributed to the cause of asthma symptoms. MicroRNAs (miRNAs) are a group of small non-coding single-stranded RNA molecules that are involved in gene silencing and posttranscriptional regulation of gene expression by targeting mRNAs. In pathological conditions, altered expression of microRNAs differentially regulates cytokines and signaling pathways and therefore, can be the underlying reason for the pathogenesis of allergic asthma. Indeed, microRNAs participate in airway inflammation via inducing airway structural cells and activating immune responses by targeting cytokines and signaling pathways. Thus, to make a complete understanding of allergic asthma, it is necessary to investigate the communication network of microRNAs with cytokines and signaling pathways which is contributed to the pathogenesis of allergic asthma. Here, we shed light on this aspect of asthma pathology by Summarizing our current knowledge of this topic.
Collapse
Affiliation(s)
- Ali Farmanzadeh
- Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
5
|
Perilla Fruit Water Extract Attenuates Inflammatory Responses and Alleviates Neutrophil Recruitment via MAPK/JNK-AP-1/c-Fos Signaling Pathway in ARDS Animal Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4444513. [PMID: 35815275 PMCID: PMC9262517 DOI: 10.1155/2022/4444513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
Airway respiratory distress syndrome (ARDS) is usually caused by a severe pulmonary infection. However, there is currently no effective treatment for ARDS. Traditional Chinese medicine (TCM) has been shown to effectively treat inflammatory lung diseases, but a clear mechanism of action of TCM is not available. Perilla fruit water extract (PFWE) has been used to treat cough, excessive mucus production, and some pulmonary diseases. Thus, we propose that PFWE may be able to reduce lung inflammation and neutrophil infiltration in a lipopolysaccharide (LPS)-stimulated murine model. C57BL/6 mice were stimulated with LPS (10 μg/mouse) by intratracheal (IT) injection and treated with three doses of PFWE (2, 5, and 8 g/kg) by intraperitoneal (IP) injections. To investigate possible mechanisms, A549 cells were treated with PFWE and stimulated with LPS. Our results showed that PFWE decreased airway resistance, neutrophil infiltration, vessel permeability, and interleukin (IL)-6 and chemokine (C-C motif) ligand 2 (CCL2/MCP-1) expressions in vivo. In addition, the PFWE inhibited the expression of IL-6, CCL2/MCP-1, chemokine (CXC motif) ligand 1 (CXCL1/GROα), and IL-8 in vitro. Moreover, PFWE also inhibited the MAPK/JNK-AP-1/c-Fos signaling pathway in A549 cells. In conclusion, we demonstrated that PFWE attenuated pro-inflammatory cytokine and chemokine levels and downregulated neutrophil recruitment through the MAPK/JNK-AP-1/c-Fos pathway. Thus, PFWE can be a potential drug to assist the treatment of ARDS.
Collapse
|
6
|
Hamed O, Joshi R, Mostafa MM, Giembycz MA. α and β Catalytic Subunits of cAMP-dependent Protein Kinase Regulate Formoterol-induced Inflammatory Gene Expression Changes in Human Bronchial Epithelial Cells. Br J Pharmacol 2022; 179:4593-4614. [PMID: 35735057 DOI: 10.1111/bph.15901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND & PURPOSE It has been proposed that genomic mechanisms contribute to the adverse-effects that are often experienced by asthmatic subjects who take regular, inhaled β2 -adrenoceptor agonists as a monotherapy. Moreover, data from preclinical models of asthma suggest that these gene expression changes are mediated by β-arrestin-2 rather than PKA. Herein, we tested this hypothesis by comparing the genomic effects of formoterol, a β2 -adrenoceptor agonist, with forskolin in human primary bronchial epithelial cells (HBEC). EXPERIMENTAL APPROACH Gene expression changes were determined by RNA-sequencing. Gene silencing and genome editing were employed to explore the roles of β-arrestin-2 and PKA. KEY RESULTS The formoterol-regulated transcriptome in HBEC treated concurrently with TNFα, was defined by 1480 unique gene expression changes. TNFα-induced transcripts modulated by formoterol were annotated with enriched gene ontology terms related to inflammation and proliferation, notably "GO:0070374~positive regulation of ERK1 and ERK2 cascade", which is an established β-arrestin-2 target. However, expression of the formoterol- and forskolin-regulated transcriptomes were highly rank-order correlated and the effects of formoterol on TNFα-induced inflammatory genes were abolished by an inhibitor of PKA. Furthermore, formoterol-induced gene expression changes in BEAS-2B bronchial epithelial cell clones deficient in β-arrestin-2 were comparable to those expressed by their parental counterparts. Contrariwise, gene expression was partially inhibited in clones lacking the α-catalytic subunit (Cα) of PKA and abolished following the additional knockdown of the β-catalytic subunit (Cβ) paralogue. CONCLUSIONS The effects of formoterol on inflammatory gene expression in airway epithelia are mediated by PKA and involve the cooperation of PKA-Cα and PKA-Cβ.
Collapse
Affiliation(s)
- Omar Hamed
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Radhika Joshi
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mahmoud M Mostafa
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark A Giembycz
- Airways Inflammation Research Group, Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Chemokines and chemokine receptors in allergic rhinitis: from mediators to potential therapeutic targets. Eur Arch Otorhinolaryngol 2022; 279:5089-5095. [PMID: 35732904 DOI: 10.1007/s00405-022-07485-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023]
Abstract
Allergic rhinitis (AR) is an immune-mediated inflammatory condition characterized by immune cell infiltration of the nasal mucosa, with symptoms of rhinorrhea, sneezing, nasal obstruction, and itchiness. Currently, common medication for AR is anti-inflammatory treatment including intranasal steroids, oral, or intranasal anti-histamines, and immunotherapy. These strategies are effective to the majority of patients with AR, but some patients under medication cannot achieve symptom relieve and suffer from bothersome side effects, indicating a demand for novel anti-inflammatory treatment as alternatives. Chemokines, a complex superfamily of small, secreted proteins, were initially recognized for their chemotactic effects on various immune cells. Chemokines constitute both physiological and inflammatory cell positioning systems and mediate cell localization to certain sites via interaction with their receptors, which are expressed on responding cells. Chemokines and their receptors participate in the sensitization, early phase response, and late phase response of AR by promoting inflammatory cell recruitment, differentiation, and allergic mediator release. In this review, we first systemically summarize chemokines and chemokine receptors that are important in AR pathophysiology and then discuss potential strategies targeting chemokines and their receptors for AR therapy.
Collapse
|
8
|
Hong JH, Lee YC. Anti-Inflammatory Effects of Cicadidae Periostracum Extract and Oleic Acid through Inhibiting Inflammatory Chemokines Using PCR Arrays in LPS-Induced Lung inflammation In Vitro. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060857. [PMID: 35743888 PMCID: PMC9225349 DOI: 10.3390/life12060857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/26/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022]
Abstract
In this study, we aimed to evaluate the anti-inflammatory effects and mechanisms of CP and OA treatments in LPS-stimulated lung epithelial cells on overall chemokines and their receptors using PCR arrays. In addition, we aimed to confirm those effects and mechanisms in LPS-stimulated lung macrophages on some chemokines and cytokines. In our study, CP treatments significantly inhibited the inflammatory mediators CCL2, CCL3, CCL4, CCL5, CCL6, CCL9, CCL11, CCL17, CCL20, CXCL1, CXCL2, CXCL3, CXCL5, CXCL7, CXCL10, TNF-α, and IL-6, while markedly suppressing NF-κB p65 nuclear translocation and the phosphorylations of PI3K p55, Akt, Erk1/2, p38, and NF-κB p65 in LPS-stimulated lung epithelial cells. CP treatments also significantly decreased the inflammatory mediators CCL2, CCL5, CCL17, CXCL1, and CXCL2, while markedly inhibiting phospho-PI3K p55 and iNOS expression in LPS-stimulated lung macrophages. Likewise, OA treatments significantly suppressed the inflammatory mediators CCL2, CCL3, CCL4, CCL5, CCL8, CCL11, CXCL1, CXCL3, CXCL5, CXCL7, CXCL10, CCRL2, TNF-α, and IL-6, while markedly reducing the phosphorylations of PI3K p85, PI3K p55, p38, JNK, and NF-κB p65 in LPS-stimulated lung epithelial cells. Finally, OA treatments significantly inhibited the inflammatory mediators CCL2, CCL5, CCL17, CXCL1, CXCL2, TNF-α, and IL-6, while markedly suppressing phospho-PI3K p55, iNOS, and Cox-2 in LPS-stimulated lung macrophages. These results prove that CP and OA treatments have anti-inflammatory effects on the inflammatory chemokines and cytokines by inhibiting pro-inflammatory mediators, including PI3K, Akt, MAPKs, NF-κB, iNOS, and Cox-2. These findings suggest that CP and OA are potential chemokine-based therapeutic substances for treating the lung and airway inflammation seen in allergic disorders.
Collapse
Affiliation(s)
| | - Young-Cheol Lee
- Correspondence: ; Tel.: +82-33-730-0672; Fax: +82-33-730-0653
| |
Collapse
|
9
|
Wang MC, Huang WC, Chen LC, Yeh KW, Lin CF, Liou CJ. Sophoraflavanone G from Sophora flavescens Ameliorates Allergic Airway Inflammation by Suppressing Th2 Response and Oxidative Stress in a Murine Asthma Model. Int J Mol Sci 2022; 23:ijms23116104. [PMID: 35682783 PMCID: PMC9181790 DOI: 10.3390/ijms23116104] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022] Open
Abstract
Sophoraflavanone G (SG), isolated from Sophora flavescens, has anti-inflammatory and anti-tumor bioactive properties. We previously showed that SG promotes apoptosis in human breast cancer cells and leukemia cells and reduces the inflammatory response in lipopolysaccharide-stimulated macrophages. We investigated whether SG attenuates airway hyper-responsiveness (AHR) and airway inflammation in asthmatic mice. We also assessed its effects on the anti-inflammatory response in human tracheal epithelial cells. Female BALB/c mice were sensitized with ovalbumin, and asthmatic mice were treated with SG by intraperitoneal injection. We also exposed human bronchial epithelial BEAS-2B cells to different concentrations of SG to evaluate its effects on inflammatory cytokine levels. SG treatment significantly reduced AHR, eosinophil infiltration, goblet cell hyperplasia, and airway inflammation in the lungs of asthmatic mice. In the lungs of ovalbumin-sensitized mice, SG significantly promoted superoxide dismutase and glutathione expression and attenuated malondialdehyde levels. SG also suppressed levels of Th2 cytokines and chemokines in lung and bronchoalveolar lavage samples. In addition, we confirmed that SG decreased pro-inflammatory cytokine, chemokine, and eotaxin expression in inflammatory BEAS-2B cells. Taken together, our data demonstrate that SG shows potential as an immunomodulator that can improve asthma symptoms by decreasing airway-inflammation-related oxidative stress.
Collapse
Affiliation(s)
- Meng-Chun Wang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan;
| | - Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan;
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (L.-C.C.); (K.-W.Y.)
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei 23656, Taiwan
| | - Li-Chen Chen
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (L.-C.C.); (K.-W.Y.)
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei 23656, Taiwan
| | - Kuo-Wei Yeh
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (L.-C.C.); (K.-W.Y.)
| | - Chwan-Fwu Lin
- Department of Cosmetic Science, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Correspondence: (C.-F.L.); (C.-J.L.); Tel.: +886-3-2118999 (ext. 5707) (C.-F.L.); +886-3-2118999 (ext. 5607) (C.-J.L.)
| | - Chian-Jiun Liou
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (L.-C.C.); (K.-W.Y.)
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan 33303, Taiwan
- Correspondence: (C.-F.L.); (C.-J.L.); Tel.: +886-3-2118999 (ext. 5707) (C.-F.L.); +886-3-2118999 (ext. 5607) (C.-J.L.)
| |
Collapse
|
10
|
RNA Sequencing Reveals the Upregulation of FOXO Signaling Pathway in Porphyromonas gingivalis Persister-Treated Human Gingival Epithelial Cells. Int J Mol Sci 2022; 23:ijms23105728. [PMID: 35628542 PMCID: PMC9146424 DOI: 10.3390/ijms23105728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Porphyromonas gingivalis as the keystone periodontopathogen plays a critical role in the pathogenesis of periodontitis, and crucially accounts for inflammatory comorbidities such as cardiovascular disease and Alzheimer's disease. We recently identified the existence of P. gingivalis persisters and revealed the unforeseen perturbation of innate response in human gingival epithelial cells (HGECs) due to these noxious persisters. Herein, RNA sequencing revealed how P. gingivalis persisters affected the expression profile of cytokine genes and related signaling pathways in HGECs. Results showed that metronidazole-treated P. gingivalis persisters (M-PgPs) impaired the innate host defense of HGECs, in a similar fashion to P. gingivalis. Notably, over one thousand differentially expressed genes were identified in HGECs treated with M-PgPs or P. gingivalis with reference to the controls. Gene Ontology and KEGG pathway analysis demonstrated significantly enriched signaling pathways, such as FOXO. Importantly, the FOXO1 inhibitor rescued the M-PgP-induced disruption of cytokine expression. This study suggests that P. gingivalis persisters may perturb innate host defense, through the upregulation of the FOXO signaling pathway. Thus, the current findings could contribute to developing new approaches to tackling P. gingivalis persisters for the effective control of periodontitis and P. gingivalis-related inflammatory comorbidities.
Collapse
|
11
|
Huang WC, Huang TH, Yeh KW, Chen YL, Shen SC, Liou CJ. Ginsenoside Rg3 ameliorates allergic airway inflammation and oxidative stress in mice. J Ginseng Res 2021; 45:654-664. [PMID: 34764720 PMCID: PMC8569325 DOI: 10.1016/j.jgr.2021.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 01/08/2021] [Accepted: 03/06/2021] [Indexed: 01/21/2023] Open
Abstract
Background Ginsenoside Rg3, isolated from Panax ginseng, has anti-inflammatory and anti-tumor activities. It is known to reduce inflammation in acute lung injury in mice, and to reduce the expression of inflammatory cytokines and COX-2 in human asthmatic airway epithelium. In this study, we attempted to determine whether ginsenoside Rg3 inhibits airway inflammation, oxidative stress, and airway hyperresponsiveness (AHR) in the lungs of asthmatic mice. We also investigated its effects on oxidative stress and the inflammatory response in tracheal epithelial cells. Methods Asthma symptoms were induced in female BALB/c mice sensitized with ovalbumin (OVA). Mice were divided into five groups: normal controls, OVA-induced asthmatic controls, and asthmatic mice treated with ginsenoside Rg3 or prednisolone by intraperitoneal injection. Inflammatory BEAS-2B cells (human tracheal epithelial cells) treated with ginsenoside Rg3 to investigate its effects on inflammatory cytokines and oxidative responses. Results Ginsenoside Rg3 treatment significantly reduced eosinophil infiltration, oxidative responses, airway inflammation, and AHR in the lungs of asthmatic mice. Ginsenoside Rg3 reduced Th2 cytokine and chemokine levels in bronchoalveolar lavage fluids and lung. Inflammatory BEAS-2B cells treated with ginsenoside Rg3 reduced the eotaxin and pro-inflammatory cytokine expressions, and monocyte adherence to BEAS-2B cells was significantly reduced as a result of decreased ICAM-1 expression. Furthermore, ginsenoside Rg3 reduced the expression of reactive oxygen species in inflammatory BEAS-2B cells. Conclusion Ginsenoside Rg3 is a potential immunomodulator that can ameliorate pathological features of asthma by decreasing oxidative stress and inflammation Ginsenoside Rg3 reduced eosinophil infiltration, and airway hyperresponsiveness in the lungs of asthmatic mice. Ginsenoside Rg3 inhibited oxidative responses in the lungs. Ginsenoside Rg3 reduced the levels of Th2 cytokines in BALF and lung. Ginsenoside Rg3 inhibited monocyte cell adherence to tracheal epithelial cells. Ginsenoside Rg3 reduced the levels of pro-inflammatory cytokines in tracheal epithelial cells.
Collapse
Affiliation(s)
- Wen-Chung Huang
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| | - Tse-Hung Huang
- Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan City, Taiwan.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Kuo-Wei Yeh
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| | - Ya-Ling Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei City, Taiwan
| | - Szu-Chuan Shen
- Graduate Program of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| | - Chian-Jiun Liou
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| |
Collapse
|
12
|
Zhu X, Zhou L, Li Q, Pan R, Zhang J, Cui Y. Combined score of C-reactive protein level and neutrophil-to-lymphocyte ratio: A novel marker in distinguishing children with exacerbated asthma. Int J Immunopathol Pharmacol 2021; 35:20587384211040641. [PMID: 34514899 PMCID: PMC8442497 DOI: 10.1177/20587384211040641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Both C-reactive protein (CRP) level and neutrophil-to-lymphocyte ratio (NLR)
are commonly elevated in patients with asthma. It is necessary to develop a
novel marker, the combined score of CRP level and NLR (C-NLR score) based on
cutoff points of CRP and NLR, and apply it in asthma diagnosis. The aim of
this study was to explore whether C-NLR could distinguish children with
exacerbated asthma. Methods Children suffering from exacerbated asthma were regarded as the asthmatic
group (n = 86), which was divided into three groups: mild
(n = 54), moderate (n = 17), and
severe (n = 15). The control group consisted of children
without any allergic disease and infection (n = 38). To
compare CRP level and NLR between the asthmatic group and control group, a
receiver-operating characteristic curve was constructed to determine area
under the curve (AUC) and optimal cutoff point. Thereafter, the C-NLR score
was classified as follows: C-NLR score of 2 with an elevated CRP level and
high NLR, a C-NLR score of 1 with one of these abnormalities, and a C-NLR
score of 0 with a normal CRP level and low NLR. The C-NLR score was then
compared among different asthma groups. Results In the control group, the CRP level and NLR were 1.9 (0.5–2.6) mg/L and 1.01
(0.69–1.31), respectively. In the asthmatic group, the CRP level and NLR
were 7.3 (3.2–14.2) mg/L and 3.08 (1.73–5.34), respectively, which were
higher than those in the control group (p < 0.001 for
CRP and p < 0.001 for NLR). The AUC of CRP was 0.86, and
the optimal cutoff point was 3.6 mg/L. The AUC of NLR was 0.86, and the
optimal cutoff point was 1.72. The AUC of the C-NLR score was 0.94, and the
optimal cutoff point was 1. Conclusions C-NLR, a novel inflammatory marker, was applied here for the exacerbated
asthma for the first time. Our study has shown C-NLR is a promising marker
to distinguish children with exacerbated asthma from healthy children.
Collapse
Affiliation(s)
- Xuming Zhu
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Lina Zhou
- Department of Respiratory Medicine, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qingqing Li
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ruilin Pan
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jian Zhang
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Yubao Cui
- Department of Clinical Laboratory, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
13
|
Busse WW, Kraft M, Rabe KF, Deniz Y, Rowe PJ, Ruddy M, Castro M. Understanding the key issues in the treatment of uncontrolled persistent asthma with type 2 inflammation. Eur Respir J 2021; 58:2003393. [PMID: 33542055 PMCID: PMC8339540 DOI: 10.1183/13993003.03393-2020] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022]
Abstract
Asthma is a complex respiratory disease that varies in severity and response to treatment. Several asthma phenotypes with unique clinical and inflammatory characteristics have been identified. Endotypes, based on distinct molecular profiles, help to further elucidate the heterogeneity within asthma. Type 2 inflammation, involving both the innate (type 2 innate lymphoid cell) and adaptive (T-helper type 2 cells) immune systems, underpins the complex pathophysiology of chronic inflammation in asthma, as well as the presence of comorbid disease (e.g. chronic rhinosinusitis with nasal polyps, allergic rhinitis and atopic dermatitis). Type 2 inflammation is characterised by upregulation of the type 2 cytokines interleukin (IL)-4, IL-5 and IL-13, IgE-mediated release of immune mediators and dysfunction of epithelial or epidermal barriers. Targeting these key proximal type 2 cytokines has shown efficacy in recent studies adopting a personalised approach to treatment using targeted biologics. Elevated levels of biomarkers downstream of type 2 cytokines, including fractional exhaled nitric oxide, serum IgE and blood and sputum eosinophils, have been linked to mechanisms involved in type 2 inflammation. They have the potential to aid diagnosis, and to predict and monitor response to treatment. The objective of this review is to summarise the current understanding of the biology of type 2 inflammation in asthma, examine its influence on type 2 inflammatory comorbidities, and discuss how type 2 inflammatory biomarkers can be harnessed to further personalise treatments in the age of biologic medicines.
Collapse
Affiliation(s)
- William W. Busse
- UW Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Monica Kraft
- University of Arizona Health Sciences Center, Tucson, AZ, USA
| | - Klaus F. Rabe
- LungenClinic Grosshansdorf (member of the German Center for Lung Research, DZL), Airway Research Center North (ARCN), Grosshansdorf, Germany
- Christian-Albrechts University (member of the German Center for Lung Research, DZL), Airway Research Center North (ARCN), Kiel, Germany
| | - Yamo Deniz
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | - Mario Castro
- University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
14
|
Ayola-Serrano NC, Roy N, Fathah Z, Anwar MM, Singh B, Ammar N, Sah R, Elba A, Utt RS, Pecho-Silva S, Rodriguez-Morales AJ, Dhama K, Quraishi S. The role of 5-lipoxygenase in the pathophysiology of COVID-19 and its therapeutic implications. Inflamm Res 2021; 70:877-889. [PMID: 34086061 PMCID: PMC8176665 DOI: 10.1007/s00011-021-01473-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/21/2021] [Accepted: 05/15/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, known as coronavirus disease 2019 (COVID-19) causes cytokine release syndrome (CRS), leading to acute respiratory distress syndrome (ARDS), acute kidney and cardiac injury, liver dysfunction, and multiorgan failure. Although several studies have discussed the role of 5-lipoxygenase (5-LOX) in viral infections, such as influenzae and SARS, it remains unexplored in the pathophysiology of COVID-19. 5-LOX acts on free arachidonic acid (AA) to form proinflammatory leukotrienes (LTs). Of note, numerous cells involved with COVID-19 (e.g., inflammatory and smooth muscle cells, platelets, and vascular endothelium) widely express leukotriene receptors. Moreover, 5-LOX metabolites induce the release of cytokines (e.g., tumour necrosis factor-α [TNF-α], interleukin-1α [IL-1α], and interleukin-1β [IL-1β]) and express tissue factor on cell membranes and activate plasmin. Since macrophages, monocytes, neutrophils, and eosinophils can express lipoxygenases, activation of 5-LOX and the subsequent release of LTs may contribute to the severity of COVID-19. This review sheds light on the potential implications of 5-LOX in SARS-CoV-2-mediated infection and the anticipated therapeutic role of 5-LOX inhibitors.
Collapse
Affiliation(s)
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| | | | - Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt
| | | | - Nour Ammar
- Department of Pediatric Dentistry and Dental Public Health, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Ranjit Sah
- Department of Microbiology, Institute of Medicine, Tribhuvan University Teaching Hospital, Kathmandu, Nepal
| | - Areej Elba
- Department of Pediatric Dentistry and Dental Public Health, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Rawan Sobhi Utt
- Faculty of Medicine, Al Quds University, Jerusalem, Palestine
| | - Samuel Pecho-Silva
- Master in Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru
- Pneumology Service, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
- Latin American Network of COVID-19 Research, Pereira, Colombia
| | - Alfonso J Rodriguez-Morales
- Master in Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru.
- Latin American Network of COVID-19 Research, Pereira, Colombia.
- Grupo de Investigacion Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| | - Sadeq Quraishi
- Department of Anesthesiology & Perioperative Medicine - Tufts Medical Center, Tufts University School of Medicine, 800 Washington St, Ziskind 6038, Boston, MA, 02111, USA
| |
Collapse
|
15
|
Pyclik MJ, Srutkova D, Razim A, Hermanova P, Svabova T, Pacyga K, Schwarzer M, Górska S. Viability Status-Dependent Effect of Bifidobacterium longum ssp . longum CCM 7952 on Prevention of Allergic Inflammation in Mouse Model. Front Immunol 2021; 12:707728. [PMID: 34354710 PMCID: PMC8329652 DOI: 10.3389/fimmu.2021.707728] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/20/2022] Open
Abstract
The classical definition of probiotics states that bacteria must be alive to be beneficial for human organism. However, recent reports show that inactivated bacteria or their effector molecules can also possess such properties. In this study, we investigated the physical and immunomodulatory properties of four Bifidobacterium strains in the heat-treated (HT) and untreated (UN) forms. We showed that temperature treatment of bacteria changes their size and charge, which affects their interaction with epithelial and immune cells. Based on the in vitro assays, we observed that all tested strains reduced the level of OVA-induced IL-4, IL-5, and IL-13 in the spleen culture of OVA-sensitized mice. We selected Bifidobacterium longum ssp. longum CCM 7952 (Bl 7952) for further analysis. In vivo experiments confirmed that untreated Bl 7952 exhibited allergy-reducing properties when administered intranasally to OVA-sensitized mice, which manifested in significant suppression of airway inflammation. Untreated Bl 7952 decreased local and systemic levels of Th2 related cytokines, OVA-specific IgE antibodies and simultaneously inhibited airway eosinophilia. In contrast, heat-treated Bl 7952 was only able to reduce IL-4 levels in the lungs and eosinophils in bronchoalveolar lavage, but increased neutrophil and macrophage numbers. We demonstrated that the viability status of Bl 7952 is a prerequisite for the beneficial effects of bacteria, and that heat treatment reduces but does not completely abolish these properties. Further research on bacterial effector molecules to elucidate the beneficial effects of probiotics in the prevention of allergic diseases is warranted.
Collapse
Affiliation(s)
- Marcelina Joanna Pyclik
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Agnieszka Razim
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Petra Hermanova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Tereza Svabova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Katarzyna Pacyga
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Novy Hradek, Czechia
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
16
|
Podlesnaja M, Pilmane M, Sumeraga G. Cytokines, Proliferation Markers, Antimicrobial Factors and Neuropeptide-Containing Innervation in Human Nasal Mucosa after Rhinoseptoplasty Procedure. Med Sci (Basel) 2021; 9:medsci9020025. [PMID: 33919374 PMCID: PMC8167725 DOI: 10.3390/medsci9020025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The nasal cavity lined by nasal mucosa, is a significant part of respiratory system of human. However, there are no studies aimed to detect a molecular phenotype of healthy and normal functioning nasal mucosa, obtained after rhinoseptoplasty procedure, to understand its physiology and growth and inflammation processes. Thus, our aim is to identify human healthy nasal mucosa cytokines, neuropeptide-containing innervation and cell proliferation markers to form a control group for further tissue investigation of human nasal polyposis as the next step of our research. The study included surgery materials from 17 healthy humans. Biotin-streptavidin immunohistochemistry was performed for detection of tissue PGP9.5, Ki-67, β-Defensin 2, IL-1, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12. Results were evaluated semi-quantitatively and by Friedman ANOVA and Spearman rang correlation tests. All factors were more widely expressed by superficial epithelium than by glandular one. Abundance of ILs-8, -10 and -12 positive cells was detected in comparison with moderate to numerous distributions of IL-1, IL-6 and β-Defensin 2. Moderate number of PGP 9.5-containing nerve fibers and only few to moderate Ki-67 positive cells were found in healthy nasal mucosa. We revealed statistically significant difference between Ki-67 and ILs-4, -6, -7, -8, -10, -12 both in healthy nasal mucosa superficial and glandular epithelium. From nasal epithelia, commonly the surface one displays more cytokines and β-Defensin 2 in comparison to the glandular one. Numerous to abundant expression of ILs-4, -6, -7, -8, -10, -12 and β-Defensin 2 in nasal superficial and glandular epithelia proves probably these factors' role into the common immune response of tissue and stimulation of immune cell differentiation.
Collapse
Affiliation(s)
- Marija Podlesnaja
- Institute of Anatomy and Anthropology, Riga Stradins University, LV-1010 Riga, Latvia;
- Correspondence:
| | - Mara Pilmane
- Institute of Anatomy and Anthropology, Riga Stradins University, LV-1010 Riga, Latvia;
| | - Gunta Sumeraga
- Department of Otorhinolaryngology, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia;
| |
Collapse
|
17
|
Sheikhpour M, Maleki M, Ebrahimi Vargoorani M, Amiri V. A review of epigenetic changes in asthma: methylation and acetylation. Clin Epigenetics 2021; 13:65. [PMID: 33781317 PMCID: PMC8008616 DOI: 10.1186/s13148-021-01049-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/04/2021] [Indexed: 12/30/2022] Open
Abstract
Several studies show that childhood and adulthood asthma and its symptoms can be modulated through epigenetic modifications. Epigenetic changes are inheritable modifications that can modify the gene expression without changing the DNA sequence. The most common epigenetic alternations consist of DNA methylation and histone modifications. How these changes lead to asthmatic phenotype or promote the asthma features, in particular by immune pathways regulation, is an understudied topic. Since external effects, like exposure to tobacco smoke, air pollution, and drugs, influence both asthma development and the epigenome, elucidating the role of epigenetic changes in asthma is of great importance. This review presents available evidence on the epigenetic process that drives asthma genes and pathways, with a particular focus on DNA methylation, histone methylation, and acetylation. We gathered and assessed studies conducted in this field over the past two decades. Our study examined asthma in different aspects and also shed light on the limitations and the important factors involved in the outcomes of the studies. To date, most of the studies in this area have been carried out on DNA methylation. Therefore, the need for diagnostic and therapeutic applications through this molecular process calls for more research on the histone modifications in this disease.
Collapse
Affiliation(s)
- Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Mobina Maleki
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Ebrahimi Vargoorani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Department of Microbiology, College of Basic Sciences, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Vahid Amiri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
18
|
From Allergy to Cancer-Clinical Usefulness of Eotaxins. Cancers (Basel) 2021; 13:cancers13010128. [PMID: 33401527 PMCID: PMC7795139 DOI: 10.3390/cancers13010128] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Eotaxins are small proteins included in the group of chemokines. They act mainly on blood cells called eosinophils which are involved in the pathogenesis of inflammatory processes. This connection leads to involvement of eotaxins in the pathogenesis of all inflammatory related diseases, such as allergic diseases and cancer. This paper summarizes the current knowledge about eotaxins, showing their usefulness as markers that can be used not only in the detection of these diseases, but also to determine the effectiveness of treatment. Abstract Eotaxins are proteins which belong to the group of cytokines. These small molecules are secreted by cells that are mainly involved in immune-mediated reactions in the course of allergic diseases. Eotaxins were discovered in 1994 and their main role was considered to be the selective recruitment of eosinophils. As those blood cells are involved in the course of all inflammatory diseases, including cancer, we decided to perform an extensive search of the literature pertaining to our investigation via the MEDLINE/PubMed database. On the basis of available literature, we can assume that eotaxins can be used as markers for the detection and determination of origin or type of allergic disease. Many publications also confirm that eotaxins can be used in the determination of allergic disease treatment. Moreover, there are also studies indicating a connection between eotaxins and cancer. Some researchers revealed that CCL11 (C-C motif chemokine ligand 11, eotaxin-1) concentrations differed between the control and tested groups indicating their possible usefulness in cancer detection. Furthermore, some papers showed usefulness of eotaxins in determining the treatment efficacy as markers of decreasing inflammation. Therefore, in this paper we present the current knowledge on eotaxins in the course of allergic and cancerous diseases.
Collapse
|
19
|
Saba E, Lee YS, Yang WK, Lee YY, Kim M, Woo SM, Kim K, Kwon YS, Kim TH, Kwak D, Park YC, Shin HJ, Han CK, Oh JW, Lee YC, Kang HS, Rhee MH, Kim SH. Effects of a herbal formulation, KGC3P, and its individual component, nepetin, on coal fly dust-induced airway inflammation. Sci Rep 2020; 10:14036. [PMID: 32820197 PMCID: PMC7441173 DOI: 10.1038/s41598-020-68965-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/25/2020] [Indexed: 02/08/2023] Open
Abstract
Coal fly dust (CFD)-induced asthma model is used as an ambient particulate matter model of serious pulmonary damage. We aimed to evaluate the effects of a combination of ginseng and Salvia plebeia R. Br extract (KGC-03-PS; KG3P) and its individual components (hispidulin, nepetin and rosmarinic acid) in a CFD-induced mouse model of airway inflammation (asthma). We also evaluated signal transduction by KG3P and its individual components in the alveolar macrophage cell line, MH-S cells. In vitro, KG3P and its individual components inhibited nitric oxide production and expression of pro-inflammatory mediators and cytokines (iNOS, COX-2, IL-1β, IL-6 and TNF-α) through the NF-κB and MAPK pathways in coal fly ash (CFA)-induced inflammation in MH-S cells. Moreover, in the CFD-induced asthma model in mice, KG3P and its predominant individual component, nepetin, inhibited Asymmetric Dimethyl arginine (ADMA) and Symmetric Dimethyl arginine (SDMA) in serum, and decreased the histopathologic score in the lungs. A significant reduction in the neutrophils and immune cells in BALF and lung tissue was demonstrated, with significant reduction in the expression of the pro-inflammatory cytokines. Finally, IRAK-1 localization was also potently inhibited by KG3P and nepetin. Thus, KG3P extract can be considered as a potent candidate for amelioration of airway inflammation.
Collapse
Affiliation(s)
- Evelyn Saba
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Young-Sil Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Dajeon, 34054, Republic of Korea
| | - Won-Kyung Yang
- Division of Respiratory Systems, Department of Internal Medicine, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea.,Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, 34520, Republic of Korea
| | - Yuan Yee Lee
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - MinKi Kim
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Su-Min Woo
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 500-757, Republic of Korea
| | - KilSoo Kim
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Young-Sam Kwon
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Tae-Hwan Kim
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dongmi Kwak
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yang-Chun Park
- Division of Respiratory Systems, Department of Internal Medicine, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Han Jae Shin
- KT&G Research Institute, Daejeon, 34128, Republic of Korea
| | - Chang Kyun Han
- KGC Research Institute, Daejeon, 34128, Republic of Korea
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Young Cheol Lee
- Department of Herbology, College of Korean Medicine, Sangji University, 83 Sangjidae-gil, Wonju, Gangwon-do, 26339, Republic of Korea
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 500-757, Republic of Korea
| | - Man Hee Rhee
- Laboratory of Physiology and Cell Signalling, Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon, 34520, Republic of Korea.
| |
Collapse
|
20
|
Zajkowska M, Mroczko B. Eotaxins and Their Receptor in Colorectal Cancer-A Literature Review. Cancers (Basel) 2020; 12:cancers12061383. [PMID: 32481530 PMCID: PMC7352276 DOI: 10.3390/cancers12061383] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies in the world, with a global incidence of almost 2 million new cases every year. Despite the availability of many diagnostic tests, including laboratory tests and molecular diagnostics, an increasing number of new cases is observed. Thus, it is very important to search new markers that would show high diagnostic sensitivity and specificity in the detection of colorectal cancer in early stages of the disease. Eotaxins are proteins that belong to the cytokine group-small molecules with a variety of applications. Their main role is the activation of basophils and eosinophils involved in inflammatory processes. Therefore, we performed an extensive search of the literature pertaining to our investigation via the MEDLINE/PubMed database. On the basis of available literature, we can assume that eotaxins accumulate in cancer cells in the course of CRC. This leads to a decrease in the chemotaxis of eosinophils, which are effector immune cells with anti-tumor activity. This may explain a decrease in their number as a defense mechanism of cancer cells against their destruction and may be useful when attempting anti-tumor therapy with the use of chemokines.
Collapse
Affiliation(s)
- Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Correspondence: ; Tel.: +48-686-5168; Fax: +48-686-5169
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland;
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
21
|
Guo X, Zheng M, Pan R, Zang B, Jin M. Hydroxysafflor Yellow A Suppresses Platelet Activating Factor-Induced Activation of Human Small Airway Epithelial Cells. Front Pharmacol 2018; 9:859. [PMID: 30123133 PMCID: PMC6085473 DOI: 10.3389/fphar.2018.00859] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
Hydroxysafflor yellow A (HSYA) is a chemical component isolated from the Chinese medicine Carthamus tinctorius L. HSYA has numerous pharmacological effects, including protecting against and mitigating some respiratory diseases such as acute lung injury and chronic obstructive pulmonary disease; however, its effect on asthma remains unclear. We previously found that HSYA attenuated ovalbumin-induced allergic asthma in guinea pigs. Platelet activating factor (PAF) is a phospholipid mediator of inflammation and an important factor in the pathological process of asthma. In this study, we investigated the anti-inflammatory effects of HSYA and its underlying mechanisms in PAF-induced human small airway epithelial cells (HSAECs). PAF-activated cells were pretreated with HSYA and/or the PAF receptor inhibitor, ginkgolide B, and we observed changes in the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha, monolayer permeability of HSAECs, and inflammatory signaling pathways. HSYA attenuated the PAF-induced increase in expression of inflammatory factors and destruction of cell-barrier function, and inhibited the expression of protein kinase C, mitogen-activated protein kinases, activator protein-1, and nuclear factor-κB activation induced by PAF. These findings suggest that HSYA may represent a potential new drug for the treatment of asthma.
Collapse
Affiliation(s)
- Xinjing Guo
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Meng Zheng
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ruiyan Pan
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Baoxia Zang
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Ming Jin
- Department of Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|