1
|
Yao CR, Jiang YX, Li SY, Lu ZJ, Long XB, Xin N, Zhang JG, Ma DD, Ying GG, Shi WJ. Evaluating pulmonary toxicity of PFOS and its alternative OBS using spheroids of A549 cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176895. [PMID: 39401594 DOI: 10.1016/j.scitotenv.2024.176895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Sodium p-perfluorous nonenoxybenzene sulfonate (OBS) is a prominent alternative to perfluorooctanesulfonic acid (PFOS). Numerous studies have demonstrated hepatotoxicity and neurotoxicity of OBS and PFOS in mammals. The lungs, as a sensitive organ, are among the potential target organs for OBS and PFOS exposure. However, their toxic effects on the lungs remain unclear. In the present study, three-dimensional (3D) spheroids constructed from A549 cells were exposed to OBS and PFOS for 7 days to evaluate pulmonary toxicity through morphological examination, growth kinetics, transcriptomic profiling, and biochemical assays. Our results showed that OBS significantly reduced the diameter, volume, and growth fraction of the spheroids compared to PFOS. Transcriptomic analysis revealed a notable enrichment of the IL-17 signaling pathway after 7 days of OBS exposure. Significant differences in the transcription of genes within this pathway were observed between OBS and PFOS exposure. OBS reduced the transcription of tnfaip3, nfkbiα, map3k8, enpp2, jun, il6, cxcl1, cxcl2, cxcl3, and cxcl8 in the IL-17 signaling pathway, while PFOS enhanced the transcription of nfkbiα. Additionally, OBS decreased the level of IL-8, whereas PFOS had a minor effect. Cluster analysis confirmed significant differences in the pulmonary toxicity between OBS and PFOS. Our study demonstrated the utility of spheroids as an in vitro cell model complemented with omics technology, for comparing the pulmonary toxicity of OBS and PFOS. It provided a novel approach for evaluating the pulmonary toxicity of new pollutants like OBS.
Collapse
Affiliation(s)
- Chong-Rui Yao
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Yu-Xia Jiang
- South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510535, China
| | - Si-Ying Li
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Zhi-Jie Lu
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Xiao-Bing Long
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Na Xin
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Jin-Ge Zhang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Dong-Dong Ma
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Wen-Jun Shi
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| |
Collapse
|
2
|
Yapto CV, Rajes K, Inselmann A, Staufenbiel S, Stolte KN, Witt M, Haag R, Dommisch H, Danker K. Topical Application of Dexamethasone-Loaded Core-Multishell Nanocarriers Against Oral Mucosal Inflammation. Macromol Biosci 2024; 24:e2400286. [PMID: 39363619 DOI: 10.1002/mabi.202400286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/14/2024] [Indexed: 10/05/2024]
Abstract
Topical treatment of oral inflammatory diseases is challenging due to the intrinsic physicochemical barriers of the mucosa and the continuous flow of saliva, which dilute drugs and limit their bioavailability. Nanocarrier technology can be an innovative approach to circumvent these problems and thus improve the efficacy of topical drug delivery to the mucosa. Core-multishell (CMS) nanocarriers are putative delivery systems with high biocompatibility and the ability to adhere to and penetrate the oral mucosa. Ester-based CMS nanocarriers release the anti-inflammatory compound dexamethasone (Dx) more efficiently than a conventional cream. Mussel-inspired functionalization of a CMS nanocarrier with catechol further improves the adhesion of the nanocarrier and may enhance the efficacy of the loaded drugs. In the present study, the properties of the ester-based CMS 10-E-15-350 nanocarrier (CMS-NC) are further evaluated in comparison to the catechol-functionalized variant (CMS-C0.08). While the mucoadhesion of CMS-NC is inhibited by saliva, CMS-C0.08 exhibits better mucoadhesion in the presence of saliva. Due to the improved adhesion properties, CMS-C0.08 loaded with dexamethasone (Dx-CMS-C0.08) shows a better anti-inflammatory effect than Dx-CMS-NC when applied dynamically. These results highlight the superiority of CMS-C0.08 over CMS-NC as an innovative drug delivery system (DDS) for the treatment of oral mucosal diseases.
Collapse
Affiliation(s)
- Cynthia V Yapto
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, 14195, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
| | - Keerthana Rajes
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, 14195, Berlin, Germany
| | - Antonia Inselmann
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
| | - Sven Staufenbiel
- Freie Universität Berlin, Institute of Pharmacy, Pharmaceutical Technology, 12169, Berlin, Germany
| | - Kim N Stolte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, 14197, Berlin, Germany
| | - Maren Witt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, 14197, Berlin, Germany
| | - Rainer Haag
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, 14195, Berlin, Germany
| | - Henrik Dommisch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, 14197, Berlin, Germany
| | - Kerstin Danker
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Berlin, Germany
| |
Collapse
|
3
|
Guerra MB, Santana KG, Momolli M, Labat R, Chavantes MC, Zammuner SR, Júnior JAS, da Palma RK, Aimbire F, de Oliveira APL. Effect of photobiomodulation in an experimental in vitro model of asthma-Copd overlap. JOURNAL OF BIOPHOTONICS 2024; 17:e202400124. [PMID: 39134306 DOI: 10.1002/jbio.202400124] [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: 03/25/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 10/10/2024]
Abstract
The objective of the study was to evaluate the effect of photobiomodulation (PBM) with laser on the inflammatory process in an experimental in vitro model of ACO. The groups were: (1) human bronchial epithelial cells (BEAS-2B); (2) BEAS-2B cells treated with dexamethasone; (3) BEAS-2B cells irradiated with laser; (4) BEAS-2B cells stimulated with cigarette smoke extract (CSE) + House Dust Mite (HDM); (5) BEAS-2B cells stimulated with CSE + HDM and treated with dexamethasone; (6) BEAS-2B cells incubated with CSE + HDM and irradiated with laser. After 24 h, cytokines were quantified. There was a reduction in TNF-α, IL-1β, IL-6, IL-4, IL-5, IL-13, IL-17, IL-21, IL-23, and an increase in IL-10 and IFN-γ in cells from the laser-irradiated ACO group compared to only ACO group. With these results, we can suggest that photobiomodulation acts in the modulation of inflammation observed in ACO, and may be a treatment option.
Collapse
Affiliation(s)
- Marina Bertoni Guerra
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Kelly Gomes Santana
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Marcos Momolli
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Rodrigo Labat
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
| | | | - Stella Regina Zammuner
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - José Antonio Silva Júnior
- Post Graduate Program in Medicine-Biophotonic, University Nove de Julho (UNINOVE), São Paulo, Brazil
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | | | - Flavio Aimbire
- Translational Medicine, Federal University of São Paulo-UNIFESP, São José dos Campos, Brazil
| | | |
Collapse
|
4
|
Ibrahim BMM, Darwish AB, Taleb SA, Mourad RM, Yassen NN, Hessin AF, Gad SA, Mohammed MA. Appraisal terpenoids rich Boswellia carterri ethyl acetate extract in binary cyclodextrin oligomer nano complex for improving respiratory distress. Sci Rep 2024; 14:16779. [PMID: 39039094 PMCID: PMC11263383 DOI: 10.1038/s41598-024-66297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 07/01/2024] [Indexed: 07/24/2024] Open
Abstract
Boswellia carterii (BC) resins plants have a long historical background as a treatment for inflammation, as indicated by information originating from multiple countries. Twenty-seven diterpenoids have been identified in ethyl acetate and total methanol BC, comprising seventeen boscartins of the cembrane-type diterpenoids and ten boscartols of the prenylaromadendrane-type diterpenoids. Moreover, twenty-one known triterpenoids have also been found, encompassing nine tirucallane-type, six ursane-type, four oleanane-type, and two lupane-type. The cembrane-type diterpenoids hold a significant position in pharmaceutical chemistry and related industries due to their captivating biological characteristics and promising pharmacological potentials. Extraction of BC, creation and assessment of nano sponges loaded with either B. carterii plant extract or DEX, are the subjects of our current investigation. With the use of ultrasound-assisted synthesis, nano sponges were produced. The entrapment efficiency (EE%) of medications in nano sponges was examined using spectrophotometry. Nano sponges were characterized using a number of methods. Within nano sponges, the EE% of medicines varied between 98.52 ± 0.07 and 99.64 ± 1.40%. The nano sponges' particle sizes varied from 105.9 ± 15.9 to 166.8 ± 26.3 nm. Drugs released from nano sponges using the Korsmeyer-Peppas concept. In respiratory distressed rats, the effects of BC plant extract, DEX salt and their nano formulations (D1, D5, P1 and P1), were tested. Treatment significantly reduced ICAM-1, LTB4, and ILβ 4 levels and improved histopathologic profiles, when compared to the positive control group. Boswellia extract and its nano sponge formulation P1 showed promising therapeutic effects. The effect of P1 may be due to synergism between both the extract and the formulation. This effect was achieved by blocking both ICAM-1 and LTB4 pathways, therefore counteracting the effects of talc powder.
Collapse
Affiliation(s)
- Bassant M M Ibrahim
- Pharmacology Department, Medical and Clinical Studies Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Asmaa Badawy Darwish
- Pharmaceutical Technology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Giza, 12622, Egypt.
| | - Sally Abou Taleb
- Pharmaceutical Technology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Giza, 12622, Egypt
| | - Reda M Mourad
- Polymers and Pigments Department, Chemical Industries Research Institute, National Research Centre, 33 El-Buhouth Street, Dokki, Giza, 12622, Egypt
| | - Noha Nazeeh Yassen
- Pathology Department, Medical and Clinical Studies Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Alyaa F Hessin
- Pharmacology Department, Medical and Clinical Studies Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Shaimaa A Gad
- Pharmacology Department, Medical and Clinical Studies Research Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Mona A Mohammed
- Pharmaceutical Technology Department, National Research Centre, 33 El-Buhouth Street, Dokki, Giza, 12622, Egypt.
- Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Egypt.
| |
Collapse
|
5
|
Shaikh TB, Chandra Y, Andugulapati SB, Sistla R. Vistusertib improves pulmonary inflammation and fibrosis by modulating inflammatory/oxidative stress mediators via suppressing the mTOR signalling. Inflamm Res 2024; 73:1223-1237. [PMID: 38789791 DOI: 10.1007/s00011-024-01894-5] [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: 03/25/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
INTRODUCTION Inflammation and oxidative stress are key factors in the development of pulmonary fibrosis (PF) by promoting the differentiation of fibroblasts through modulating various pathways including Wnt/β-catenin, TGF-β and mTOR signalling. OBJECTIVE AND METHODS This study aimed to evaluate the effects and elucidate the mechanisms of vistusertib (VSB) in treating pulmonary inflammation/fibrosis, specifically by targeting the mTOR pathway using various in vitro and in vivo models. RESULTS Lipopolysaccharide (LPS)-induced inflammation model in macrophages (RAW 264.7), epithelial (BEAS-2B) and endothelial (HMVEC-L) cells revealed that treatment with VSB significantly reduced the IL-6, TNF-α, CCL2, and CCL7 expression. TGF-β induced differentiation was also significantly reduced upon VSB treatment in fibrotic cells (LL29 and DHLF). Further, bleomycin-induced inflammation and fibrosis models demonstrated that treatment with VSB significantly ameliorated the severe inflammation, and lung architectural distortion, by reducing the inflammatory markers expression/levels, inflammatory cells and oxidative stress indicators. Further, fibrosis model results exhibited that, VSB treatment significantly reduced the α-SMA, collagen and TGF-β expressions, improved the lung architecture and restored lung functions. CONCLUSION Overall, this study uncovers the anti-inflammatory/anti-fibrotic effects of VSB by modulating the mTOR activation. Although VSB was tested for lung fibrosis, it can be tested for other fibrotic disorders to improve the patient's survival and quality of life.
Collapse
Affiliation(s)
- Taslim B Shaikh
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India
| | - Yogesh Chandra
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India
| | - Sai Balaji Andugulapati
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| | - Ramakrishna Sistla
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201 002, India.
| |
Collapse
|
6
|
Suh DI, Johnston SL. The Wiser Strategy of Using Beta-Agonists in Asthma: Mechanisms and Rationales. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:217-234. [PMID: 38910281 PMCID: PMC11199159 DOI: 10.4168/aair.2024.16.3.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Concerns regarding the safety of beta-2 agonists have led to revisions of the major asthma guidelines to better address these issues. Although these updates allow for a combination of previous and current strategies, they may confuse clinical practitioners. Beta-2 agonists are vital for alleviating asthma symptoms by relaxing smooth muscles; however, they also pose significant risks by inducing pro-inflammatory mediators both in vitro and in vivo. In addition to the risks of overuse and symptom masking, the use of beta-agonists alone at therapeutic doses can worsen airway inflammation and enhance virus-induced inflammation during asthma exacerbation. Inhaled corticosteroids (ICS) can effectively prevent these adverse effects. With new insights into the mechanisms of these adverse events, reserving short-acting beta-agonists for acute symptom relief during exacerbations and only for those who are already on ICS or oral steroids represents a careful approach to using beta-agonists with least adverse effects in patients with asthma. However, a major drawback of this approach is the potential non-compliance with ICS, leading to beta-agonist use without the necessary counteraction by ICS. An optimal strategy, both during and outside exacerbations, would integrate beta-agonists into an anti-inflammatory regimen that includes ICS, ideally combined with the same inhaler to ensure their concurrent use where finances allow. This would maintain the beneficial effects of beta-agonists, such as bronchodilation, while preventing the adverse effects from the induction of inflammatory mediators. This method is aligned with diverse clinical settings, maximizes the safe use of beta-agonists, and supports a comprehensive guideline-compliant management strategy.
Collapse
Affiliation(s)
- Dong In Suh
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Pediatrics, Seoul National University Hospital, Seoul, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | | |
Collapse
|
7
|
Yang Z, Man J, Liu Y, Zhang H, Wu D, Shao D, Hao B, Wang S. Study on the Alleviating Effect and Potential Mechanism of Ethanolic Extract of Limonium aureum (L.) Hill. on Lipopolysaccharide-Induced Inflammatory Responses in Macrophages. Int J Mol Sci 2023; 24:16272. [PMID: 38003461 PMCID: PMC10671607 DOI: 10.3390/ijms242216272] [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: 10/23/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammation is the host response of immune cells during infection and traumatic tissue injury. An uncontrolled inflammatory response leads to inflammatory cascade, which in turn triggers a variety of diseases threatening human and animal health. The use of existing inflammatory therapeutic drugs is constrained by their high cost and susceptibility to systemic side effects, and therefore new therapeutic candidates for inflammatory diseases need to be urgently developed. Natural products are characterized by wide sources and rich pharmacological activities, which are valuable resources for the development of new drugs. This study aimed to uncover the alleviating effect and potential mechanism of natural product Limonium aureum (LAH) on LPS-induced inflammatory responses in macrophages. The experimental results showed that the optimized conditions for LAH ultrasound-assisted extraction via response surface methodology were an ethanol concentration of 72%, a material-to-solvent ratio of 1:37 g/mL, an extraction temperature of 73 °C, and an extraction power of 70 W, and the average extraction rate of LAH total flavonoids was 0.3776%. Then, data of 1666 components in LAH ethanol extracts were obtained through quasi-targeted metabolomics analysis. The ELISA showed that LAH significantly inhibited the production of pro-inflammatory cytokines while promoting the secretion of anti-inflammatory cytokines. Finally, combined with the results of network pharmacology analysis and protein expression validation of hub genes, it was speculated that LAH may alleviate LPS-induced inflammatory responses of macrophages through the AKT1/RELA/PTGS2 signaling pathway and the MAPK3/JUN signaling pathway. This study preliminarily revealed the anti-inflammatory activity of LAH and the molecular mechanism of its anti-inflammatory action, and provided a theoretical basis for the development of LAH as a new natural anti-inflammatory drug.
Collapse
Affiliation(s)
- Zhen Yang
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Jingyuan Man
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Yu Liu
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Hongjuan Zhang
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Di Wu
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Dan Shao
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Baocheng Hao
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| | - Shengyi Wang
- Key Laboratory of New Animal Drug Project, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Z.Y.); (Y.L.); (H.Z.); (D.W.); (D.S.)
| |
Collapse
|
8
|
Li JX, Huang XZ, Fu WP, Zhang XH, Mauki DH, Zhang J, Sun C, Dai LM, Zhong L, Yu L, Zhang YP. Remote regulation of rs80245547 and rs72673891 mediated by transcription factors C-Jun and CREB1 affect GSTCD expression. iScience 2023; 26:107383. [PMID: 37609638 PMCID: PMC10440715 DOI: 10.1016/j.isci.2023.107383] [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: 06/16/2022] [Revised: 12/30/2022] [Accepted: 07/11/2023] [Indexed: 08/24/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), the third leading cause of death worldwide, is influenced by genetic factors. The genetic signal rs10516526 in the glutathione S-transferase C-terminal domain containing (GSTCD) gene is a highly significant and reproducible signal associated with lung function and COPD on chromosome 4q24. In this study, comprehensive bioinformatics analyses and experimental verifications were detailly implemented to explore the regulation mechanism of rs10516526 and GSTCD in COPD. The results suggested that low expression of GSTCD was associated with COPD (p = 0.010). And C-Jun and CREB1 transcription factors were found to be essential for the regulation of GSTCD by rs80245547 and rs72673891. Moreover, rs80245547T and rs72673891G had a stronger binding ability to these transcription factors, which may promote the allele-specific long-range enhancer-promoter interactions on GSTCD, thus making COPD less susceptible. Our study provides a new insight into the relationship between rs10516526, GSTCD, and COPD.
Collapse
Affiliation(s)
- Jin-Xiu Li
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650000, China
| | - Xue-Zhen Huang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - Wei-ping Fu
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Xiao-hua Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - David H. Mauki
- Faculty of Pharmaceutical Sciences, Institute of Biomedicine and Biotechnology, Center for Cancer Immunology, Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Shenzhen 518000, Guangdong China
| | - Jing Zhang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Chang Sun
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- College of Life Sciences, Shaanxi Normal University, Xi’an 710000, China
| | - Lu-Ming Dai
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Li Zhong
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- College of Life Sciences, Shaanxi Normal University, Xi’an 710000, China
- Provincial Demonstration Center for Experimental Biology Education, Shaanxi Normal University, Xi’an 710000, China
| | - Li Yu
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - Ya-ping Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650000, China
| |
Collapse
|
9
|
Ambwani S, Dolma R, Sharma R, Kaur A, Singh H, Ruj A, Ambwani TK. Modulation of inflammatory and oxidative stress biomarkers due to dexamethasone exposure in chicken splenocytes. Vet Immunol Immunopathol 2023; 262:110632. [PMID: 37517103 DOI: 10.1016/j.vetimm.2023.110632] [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: 05/26/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Dexamethasone (DEXA) is a potent corticosteroid, commonly used for treating inflammatory, hypersensitive and allergic conditions. It is administered to birds with tumours. Many studies were conducted on its immunosuppressive effects; however none of the similar study is available employing chicken splenocytes culture system. The present study was conducted to assess DEXA induced alterations in inflammatory and oxidative stress biomarkers in chicken splenocytes due to its in vitro exposure. The maximum non-cytotoxic dose (MNCD) was evaluated and was further used for conducting lymphocytes proliferation assay (LPA), antioxidant assays (lipid peroxidation, GSH, superoxide dismutase and nitric oxide assays) and assessment of mRNA levels of various genes (IL-1β, IL-6, IL-10, LITAF, iNOS, NF-κB1, Nrf-2, Caspase-3 and -9) through qPCR. The MNCD was determined to be 30 ng/ml in chicken splenocytes culture system. DEXA caused reduction in B and T lymphocytes proliferation indicating its immunosuppressive effects, however improved the antioxidant status of the exposed splenocytes. The expression levels of IL-1β, IL-6, iNOS, LITAF and NF-κB1 were significantly reduced while IL-10 was enhanced, which signify potent anti-inflammatory potential of DEXA. NF-κB is a major transcription factor that regulates genes responsible for both, innate and adaptive immune responses and elicits inflammation. The nuclear factor erythroid 2-related factor 2 (Nrf-2) level was found to be up-regulated. Nrf-2 plays important role in combating the oxidant stress and its increased expression could be the reason of improved antioxidant status of DEXA exposed cells. Present findings indicated that DEXA exhibited modulation in anti-inflammatory, immunomodulatory and antioxidant mediators in chicken splenocytes.
Collapse
Affiliation(s)
- Sonu Ambwani
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India.
| | - Rigzin Dolma
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| | - Raunak Sharma
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| | - Amandip Kaur
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| | - Himani Singh
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| | - Anamitra Ruj
- Department of Molecular Biology and Genetic Engineering, C.B.S.H., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| | - Tanuj Kumar Ambwani
- Department of Veterinary Physiology and Biochemistry, C.V.A.S., Govind Ballabh Pant University of Agriculture & Technology, Pantnagar 263145, Uttarakhand, India
| |
Collapse
|
10
|
Sung S, Steele LA, Risser GE, Spiller KL. Biomaterial-Assisted Macrophage Cell Therapy for Regenerative Medicine. Adv Drug Deliv Rev 2023:114979. [PMID: 37394101 DOI: 10.1016/j.addr.2023.114979] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Although most tissue types are capable of some form of self-repair and regeneration, injuries that are larger than a critical threshold or those occurring in the setting of certain diseases can lead to impaired healing and ultimately loss of structure and function. The immune system plays an important role in tissue repair and must be considered in the design of therapies in regenerative medicine. In particular, macrophage cell therapy has emerged as a promising strategy that leverages the reparative roles of these cells. Macrophages are critical for successful tissue repair and accomplish diverse functions throughout all phases of the process by dramatically shifting in phenotypes in response to microenvironmental cues. Depending on their response to various stimuli, they may release growth factors, support angiogenesis, and facilitate extracellular matrix remodeling. However, this ability to rapidly shift phenotype is also problematic for macrophage cell therapy strategies, because adoptively transferred macrophages fail to maintain therapeutic phenotypes following their administration to sites of injury or inflammation. Biomaterials are a potential way to control macrophage phenotype in situ while also enhancing their retention at sites of injury. Cell delivery systems incorporated with appropriately designed immunomodulatory signals have potential to achieve tissue regeneration in intractable injuries where traditional therapies have failed. Here we explorecurrent challenges in macrophage cell therapy, especially retention and phenotype control, how biomaterials may overcome them, and opportunities for next generation strategies. Biomaterials will be an essential tool to advance macrophage cell therapy for widespread clinical applications.
Collapse
Affiliation(s)
- Samuel Sung
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Lindsay A Steele
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Gregory E Risser
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
11
|
Jain N, Zhao Z, Feucht J, Koche R, Iyer A, Dobrin A, Mansilla-Soto J, Yang J, Zhan Y, Lopez M, Gunset G, Sadelain M. TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature 2023; 615:315-322. [PMID: 36755094 PMCID: PMC10511001 DOI: 10.1038/s41586-022-05692-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/30/2022] [Indexed: 02/10/2023]
Abstract
Further advances in cell engineering are needed to increase the efficacy of chimeric antigen receptor (CAR) and other T cell-based therapies1-5. As T cell differentiation and functional states are associated with distinct epigenetic profiles6,7, we hypothesized that epigenetic programming may provide a means to improve CAR T cell performance. Targeting the gene that encodes the epigenetic regulator ten-eleven translocation 2 (TET2)8 presents an interesting opportunity as its loss may enhance T cell memory9,10, albeit not cause malignancy9,11,12. Here we show that disruption of TET2 enhances T cell-mediated tumour rejection in leukaemia and prostate cancer models. However, loss of TET2 also enables antigen-independent CAR T cell clonal expansions that may eventually result in prominent systemic tissue infiltration. These clonal proliferations require biallelic TET2 disruption and sustained expression of the AP-1 factor BATF3 to drive a MYC-dependent proliferative program. This proliferative state is associated with reduced effector function that differs from both canonical T cell memory13,14 and exhaustion15,16 states, and is prone to the acquisition of secondary somatic mutations, establishing TET2 as a guardian against BATF3-induced CAR T cell proliferation and ensuing genomic instability. Our findings illustrate the potential of epigenetic programming to enhance T cell immunity but highlight the risk of unleashing unchecked proliferative responses.
Collapse
Affiliation(s)
- Nayan Jain
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Zeguo Zhao
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Judith Feucht
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- University Children's Hospital, Tübingen, Germany
| | - Richard Koche
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Archana Iyer
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Anton Dobrin
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Jorge Mansilla-Soto
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Julie Yang
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Yingqian Zhan
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Michael Lopez
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Gertrude Gunset
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Michel Sadelain
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA.
| |
Collapse
|
12
|
The Preventive Effect of Specific Collagen Peptides against Dexamethasone-Induced Muscle Atrophy in Mice. Molecules 2023; 28:molecules28041950. [PMID: 36838938 PMCID: PMC9960993 DOI: 10.3390/molecules28041950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Muscle atrophy, also known as muscle wasting, is the thinning of muscle mass due to muscle disuse, aging, or diseases such as cancer or neurological problems. Muscle atrophy is closely related to the quality of life and has high morbidity and mortality. However, therapeutic options for muscle atrophy are limited, so studies to develop therapeutic agents for muscle loss are always required. For this study, we investigated how orally administered specific collagen peptides (CP) affect muscle atrophy and elucidated its molecular mechanism using an in vivo model. We treated mice with dexamethasone (DEX) to induce a muscular atrophy phenotype and then administered CP (0.25 and 0.5 g/kg) for four weeks. In a microcomputed tomography analysis, CP (0.5 g/kg) intake significantly increased the volume of calf muscles in mice with DEX-induced muscle atrophy. In addition, the administration of CP (0.25 and 0.5 g/kg) restored the weight of the gluteus maximus and the fiber cross-sectional area (CSA) of the pectoralis major and calf muscles, which were reduced by DEX. CP significantly inhibited the mRNA expression of myostatin and the phosphorylation of Smad2, but it did not affect TGF-β, BDNF, or FNDC5 gene expression. In addition, AKT/mTOR, a central pathway for muscle protein synthesis and related to myostatin signaling, was enhanced in the groups that were administered CP. Finally, CP decreased serum albumin levels and increased TNF-α gene expression. Collectively, our in vivo results demonstrate that CP can alleviate muscle wasting through a multitude of mechanisms. Therefore, we propose CP as a supplement or treatment to prevent muscle atrophy.
Collapse
|
13
|
Phytocannabinoids Act Synergistically with Non-Steroidal Anti-Inflammatory Drugs Reducing Inflammation in 2D and 3D In Vitro Models. Pharmaceuticals (Basel) 2022; 15:ph15121559. [PMID: 36559009 PMCID: PMC9787964 DOI: 10.3390/ph15121559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Lung inflammation is associated with elevated pro-inflammatory cytokines and chemokines. Treatment with FCBD:std (standard mix of cannabidiol [CBD], cannabigerol [CBG] and tetrahydrocannabivarin [THCV]) leads to a marked reduction in the inflammation of alveolar epithelial cells, but not in macrophages. In the present study, the combined anti-inflammatory effect of FCBD:std with two corticosteroids (dexamethasone and budesonide) and two non-steroidal anti-inflammatory drugs (NSAID; ibuprofen and diclofenac), was examined. Enzyme-linked immunosorbent assay (ELISA) was used to determine protein levels. Gene expression was determined by quantitative real-time PCR. Inhibition of cyclo-oxygenase (COX) activity was determined in vitro. FCBD:std and diclofenac act synergistically, reducing IL-8 levels in macrophages and lung epithelial cells. FCBD:std plus diclofenac also reduced IL-6, IL-8 and CCL2 expression levels in co-cultures of macrophages and lung epithelial cells, in 2D and 3D models. Treatment by FCBD:std and/or NSAID reduced COX-1 and COX-2 gene expression but not their enzymatic activity. FCBD:std and diclofenac exhibit synergistic anti-inflammatory effects on macrophages and lung epithelial cells, yet this combined activity needs to be examined in pre-clinical studies and clinical trials.
Collapse
|
14
|
Park JW, Choi J, Lee J, Park JM, Kim SM, Min JH, Seo DY, Goo SH, Kim JH, Kwon OK, Lee K, Ahn KS, Oh SR, Lee JW. Methyl P-Coumarate Ameliorates the Inflammatory Response in Activated-Airway Epithelial Cells and Mice with Allergic Asthma. Int J Mol Sci 2022; 23:ijms232314909. [PMID: 36499236 PMCID: PMC9736825 DOI: 10.3390/ijms232314909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/03/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Methyl p-coumarate (methyl p-hydroxycinnamate) (MH) is a natural compound found in a variety of plants. In the present study, we evaluated the ameliorative effects of MH on airway inflammation in an experimental model of allergic asthma (AA). In this in vitro study, MH was found to exert anti-inflammatory activity on PMA-stimulated A549 airway epithelial cells by suppressing the secretion of IL-6, IL-8, MCP-1, and ICAM-1. In addition, MH exerted an inhibitory effect not only on NF-κB (p-NF-κB and p-IκB) and AP-1 (p-c-Fos and p-c-Jun) activation but also on A549 cell and EOL-1 cell (eosinophil cell lines) adhesion. In LPS-stimulated RAW264.7 macrophages, MH had an inhibitory effect on TNF-α, IL-1β, IL-6, and MCP-1. The results from in vivo study revealed that the increases in eosinophils/Th2 cytokines/MCP-1 in the bronchoalveolar lavage fluid (BALF) and IgE in the serum of OVA-induced mice with AA were effectively inhibited by MH administration. MH also exerted a reductive effect on the immune cell influx, mucus secretion, and iNOS/COX-2 expression in the lungs of mice with AA. The effects of MH were accompanied by the inactivation of NF-κB. Collectively, the findings of the present study indicated that MH attenuates airway inflammation in mice with AA, suggesting its potential as an adjuvant in asthma therapy.
Collapse
Affiliation(s)
- Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jinseon Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin-Mi Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Seong-Man Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jae-Hong Min
- Laboratory Animal Resources Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Osong Health Technology Administration Complex, Cheongju 28159, Republic of Korea
| | - Da-Yun Seo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Soo-Hyeon Goo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Ju-Hee Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheonju 28116, Republic of Korea
| | - Kihoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheonju 28116, Republic of Korea
- Correspondence: (S.-R.O.); (J.-W.L.)
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- Correspondence: (S.-R.O.); (J.-W.L.)
| |
Collapse
|
15
|
Effect of an experimental desensitizing gel on bleaching-induced tooth sensitivity after in-office bleaching—a double-blind, randomized controlled trial. Clin Oral Investig 2022; 27:1567-1576. [PMID: 36418502 PMCID: PMC9685084 DOI: 10.1007/s00784-022-04778-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/06/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To evaluate the risk and intensity of tooth sensitivity (TS), and the efficacy of in-office bleaching after applying an experimental desensitizing gel composed of 10% calcium gluconate, 0.1% dexamethasone acetate, 10% potassium nitrate, and 5% glutaraldehyde. MATERIAL AND METHODS In a split-mouth, double-blind, placebo-controlled study, 50 participants had their upper hemiarches randomized into experimental and placebo groups. Desensitizing and placebo gels were applied for 10 min before in-office bleaching (35% hydrogen peroxide, 1 × 50 min; two bleaching sessions; 1-week interval). TS was recorded immediately after bleaching, 1, 24, and 48 h after each session, with a 0-10 visual analogue scale (VAS) and a five-point numerical rating scale (NRS). The color was recorded in all groups at baseline, 1 week after each session, and 1 month after the end of bleaching using shade guide units (ΔSGUs) and a spectrophotometer (ΔEab, ΔE00, and ΔWID). RESULTS Most participants (96%) felt some discomfort during treatment regardless of the study group. The odds ratio for pain was 0.65 (95% CI 0.1 to 4.1; p = 1.0). The intensity of TS did not differ between groups (p > 0.31), and it was only 0.34 VAS units lower in the experimental group. A significant color change occurred in both groups regardless of the group. CONCLUSIONS The desensitizing experimental gel applied before in-office bleaching did not reduce the risk and the intensity of TS and did not affect color change. CLINICAL RELEVANCE Although the experimental desensitizing agent with varying mechanisms of action did not jeopardize the color change, it did not reduce the risk or intensity of in-office bleaching. CLINICAL TRIAL REGISTRATION NUMBER RBR-7T7D4D.
Collapse
|
16
|
Marhuenda E, Villarino A, Narciso M, Elowsson L, Almendros I, Westergren-Thorsson G, Farré R, Gavara N, Otero J. Development of a physiomimetic model of acute respiratory distress syndrome by using ECM hydrogels and organ-on-a-chip devices. Front Pharmacol 2022; 13:945134. [PMID: 36188621 PMCID: PMC9517737 DOI: 10.3389/fphar.2022.945134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Acute Respiratory Distress Syndrome is one of the more common fatal complications in COVID-19, characterized by a highly aberrant inflammatory response. Pre-clinical models to study the effect of cell therapy and anti-inflammatory treatments have not comprehensively reproduced the disease due to its high complexity. This work presents a novel physiomimetic in vitro model for Acute Respiratory Distress Syndrome using lung extracellular matrix-derived hydrogels and organ-on-a-chip devices. Monolayres of primary alveolar epithelial cells were cultured on top of decellullarized lung hydrogels containing primary lung mesenchymal stromal cells. Then, cyclic stretch was applied to mimic breathing, and an inflammatory response was induced by using a bacteriotoxin hit. Having simulated the inflamed breathing lung environment, we assessed the effect of an anti-inflammatory drug (i.e., dexamethasone) by studying the secretion of the most relevant inflammatory cytokines. To better identify key players in our model, the impact of the individual factors (cyclic stretch, decellularized lung hydrogel scaffold, and the presence of mesenchymal stromal cells) was studied separately. Results showed that developed model presented a more reduced inflammatory response than traditional models, which is in line with what is expected from the response commonly observed in patients. Further, from the individual analysis of the different stimuli, it was observed that the use of extracellular matrix hydrogels obtained from decellularized lungs had the most significant impact on the change of the inflammatory response. The developed model then opens the door for further in vitro studies with a better-adjusted response to the inflammatory hit and more robust results in the test of different drugs or cell therapy.
Collapse
Affiliation(s)
- Esther Marhuenda
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Alvaro Villarino
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
| | - Maria Narciso
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Linda Elowsson
- Lung Biology, Biomedical Center, Department of Medical Science,Lund University, Lund, Sweden
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Núria Gavara
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
- *Correspondence: Jorge Otero,
| |
Collapse
|
17
|
Wu Q, Pennini ME, Bergmann JN, Kozak ML, Herring K, Sciarretta KL, Armstrong KL. Applying lessons learned from COVID-19 therapeutic trials to improve future ALI/ARDS trials. Open Forum Infect Dis 2022; 9:ofac381. [PMID: 35983268 PMCID: PMC9379817 DOI: 10.1093/ofid/ofac381] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Host-directed therapeutics targeting immune dysregulation are considered the most promising approach to address the unmet clinical need for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) related to coronavirus disease 2019 (COVID-19). To better understand the current clinical study landscape and gaps in treating hospitalized patients with severe or critical COVID-19, we identified COVID-19 trials developing host-directed therapies registered at ClinicalTrials.gov and discussed the factors contributing to the success vs failure of these studies. We have learned, instead of the one-size-fits-all approach, future clinical trials evaluating a targeted immunomodulatory agent in heterogeneous patients with ALI/ARDS due to COVID-19 or other infectious diseases can use immune-based biomarkers in addition to clinical and demographic characteristics to improve patient stratification and inform clinical decision-making. Identifying distinct patient subgroups based on immune profiles across the disease trajectory, regardless of the causative pathogen, may accelerate evaluating host-directed therapeutics in trials of ALI/ARDS and related conditions (eg, sepsis).
Collapse
Affiliation(s)
- Qun Wu
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Meghan E Pennini
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Julie N Bergmann
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Marina L Kozak
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kristen Herring
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Sciarretta
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Armstrong
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| |
Collapse
|
18
|
Nanoparticle-mediated corneal neovascularization treatments: Toward new generation of drug delivery systems. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.06.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Muhammad W, Zhu J, Zhai Z, Xie J, Zhou J, Feng X, Feng B, Pan Q, Li S, Venkatesan R, Li P, Cao H, Gao C. ROS-responsive polymer nanoparticles with enhanced loading of dexamethasone effectively modulate the lung injury microenvironment. Acta Biomater 2022; 148:258-270. [PMID: 35724918 PMCID: PMC9212862 DOI: 10.1016/j.actbio.2022.06.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022]
Abstract
The acute lung injury (ALI) is an inflammatory disorder associated with cytokine storm, which activates various reactive oxygen species (ROS) signaling pathways and causes severe complications in patients as currently seen in coronavirus disease 2019 (COVID-19). There is an urgent need for medication of the inflammatory lung environment and effective delivery of drugs to lung to reduce the burden of high doses of medications and attenuate inflammatory cells and pathways. Herein, we prepared dexamethasone-loaded ROS-responsive polymer nanoparticles (PFTU@DEX NPs) by a modified emulsion approach, which achieved high loading content of DEX (11.61 %). DEX was released faster from the PFTU@DEX NPs in a ROS environment, which could scavenge excessive ROS efficiently both in vitro and in vivo. The PFTU NPs and PFTU@DEX NPs showed no hemolysis and cytotoxicity. Free DEX, PFTU NPs and PFTU@DEX NPs shifted M1 macrophages to M2 macrophages in RAW264.7 cells, and showed anti-inflammatory modulation to A549 cells in vitro. The PFTU@DEX NPs treatment significantly reduced the increased total protein concentration in BALF of ALI mice. The delivery of PFTU@DEX NPs decreased the proportion of neutrophils significantly, mitigated the cell apoptosis remarkably compared to the other groups, reduced M1 macrophages and increased M2 macrophages in vivo. Moreover, the PFTU@DEX NPs had the strongest ability to suppress the expression of NLRP3, Caspase1, and IL-1β. Therefore, the PFTU@DEX NPs could efficiently suppress inflammatory cells, ROS signaling pathways, and cell apoptosis to ameliorate LPS-induced ALI. STATEMENT OF SIGNIFICANCE: The acute lung injury (ALI) is an inflammatory disorder associated with cytokine storm, which activates various reactive oxygen species (ROS) signaling pathways and causes severe complications in patients. There is an urgent need for medication of the inflammatory lung environment and effective delivery of drugs to modulate the inflammatory disorder and suppress the expression of ROS and inflammatory cytokines. The inhaled PFTU@DEX NPs prepared through a modified nanoemulsification method suppressed the activation of NLRP3, induced the polarization of macrophage phenotype from M1 to M2, and thereby reduced the neutrophil infiltration, inhibited the release of proteins and inflammatory mediators, and thus decreased the acute lung injury in vivo.
Collapse
Affiliation(s)
- Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiaqi Zhu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiahang Zhou
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Rajiu Venkatesan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Pan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
20
|
Caramori G, Nucera F, Mumby S, Lo Bello F, Adcock IM. Corticosteroid resistance in asthma: Cellular and molecular mechanisms. Mol Aspects Med 2022; 85:100969. [PMID: 34090658 DOI: 10.1016/j.mam.2021.100969] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/17/2022]
Abstract
Inhaled glucocorticoids (GCs) are drugs widely used as treatment for asthma patients. They prevent the recruitment and activation of lung immune and inflammatory cells and, moreover, have profound effects on airway structural cells to reverse the effects of disease on airway inflammation. GCs bind to a specific receptor, the glucocorticoid receptor (GR), which is a member of the nuclear receptor superfamily and modulates pro- and anti-inflammatory gene transcription through a number of distinct and complementary mechanisms. Targets genes include many pro-inflammatory mediators such as chemokines, cytokines, growth factors and their receptors. Inhaled GCs are very effective for most asthma patients with little, if any, systemic side effects depending upon the dose. However, some patients show poor asthma control even after the administration of high doses of topical or even systemic GCs. Several mechanisms relating to inflammation have been considered to be responsible for the onset of the relative GC resistance observed in these patients. In these patients, the side-effect profile of GCs prevent continued use of high doses and new drugs are needed. Targeting the defective pathways associated with GC function in these patients may also reactivate GC responsiveness.
Collapse
Affiliation(s)
- Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy.
| | - Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London and the NIHR Imperial Biomedical Research Centre, London, UK
| | - Federica Lo Bello
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London and the NIHR Imperial Biomedical Research Centre, London, UK.
| |
Collapse
|
21
|
Zhu T, Brown AP, Cai LP, Quon G, Ji H. Single-Cell RNA-Seq Analysis Reveals Lung Epithelial Cell Type-Specific Responses to HDM and Regulation by Tet1. Genes (Basel) 2022; 13:genes13050880. [PMID: 35627266 PMCID: PMC9140484 DOI: 10.3390/genes13050880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Tet1 protects against house dust mite (HDM)-induced lung inflammation in mice and alters the lung methylome and transcriptome. In order to explore the role of Tet1 in individual lung epithelial cell types in HDM-induced inflammation, we established a model of HDM-induced lung inflammation in Tet1 knockout and littermate wild-type mice, then studied EpCAM+ lung epithelial cells using single-cell RNA-seq analysis. We identified eight EpCAM+ lung epithelial cell types, among which AT2 cells were the most abundant. HDM challenge altered the relative abundance of epithelial cell types and resulted in cell type-specific transcriptomic changes. Bulk and cell type-specific analysis also showed that loss of Tet1 led to the altered expression of genes linked to augmented HDM-induced lung inflammation, including alarms, detoxification enzymes, oxidative stress response genes, and tissue repair genes. The transcriptomic regulation was accompanied by alterations in TF activities. Trajectory analysis supports that HDM may enhance the differentiation of AP and BAS cells into AT2 cells, independent of Tet1. Collectively, our data showed that lung epithelial cells had common and unique transcriptomic signatures of allergic lung inflammation. Tet1 deletion altered transcriptomic networks in various lung epithelial cells, which may promote allergen-induced lung inflammation.
Collapse
Affiliation(s)
- Tao Zhu
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Anthony P. Brown
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Lucy P. Cai
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
| | - Gerald Quon
- Department of Molecular and Cellular Biology, Genome Center, University of California, Davis, CA 95616, USA;
| | - Hong Ji
- California National Primate Research Center, University of California, Davis, CA 95616, USA; (T.Z.); (A.P.B.); (L.P.C.)
- Department of Anatomy, Physiology and Cell biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-530-754-0679
| |
Collapse
|
22
|
Mechanism of Peitu Shengjin Formula Shenlingbaizhu Powder in Treating Bronchial Asthma and Allergic Colitis through Different Diseases with Simultaneous Treatment Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4687788. [PMID: 35586697 PMCID: PMC9110165 DOI: 10.1155/2022/4687788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/08/2022] [Accepted: 03/26/2022] [Indexed: 11/18/2022]
Abstract
Background Shenlingbaizhu powder (SLBZP), one of the classic Earth-cultivating and gold-generating prescriptions of traditional Chinese medicine, is widely used to treat various diseases. However, the pharmacological mechanisms of SLBZP on bronchial asthma (BA) and allergic colitis (AC) remain to be elucidated. Methods Network pharmacology and molecular docking technology were used to explore the potential mechanism of SLBZP in treating BA and AC with the simultaneous treatment of different diseases. The potential active compounds of SLBZP and their corresponding targets were obtained from BATMAN-TCM, ETCM, SymMap TCM@TAIWAN, and TCMSP databases. BA and AC disease targets were collected through DisGeNET, TTD, GeneCards, PharmGKB, OMIM, NCBI, The Human Phenotype Ontology, and DrugBank databases. Common targets for drugs and diseases were screened by using the bioinformatics and evolutionary genomics platform. The analyses and visualizations of Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of common targets were carried out by R software. The key targets were screened by using the plug-in “cytoHubba” of Cytoscape software, and the “active compound-key target” network was constructed. Molecular docking analysis was performed using AutoDock software. The miRTarBase database was used to predict microRNAs (miRNAs) targeting key targets, and the key target-miRNA network was constructed. Result Through screening, 246 active compounds and 281 corresponding targets were obtained. Common targets were mainly enriched in 2933 biological processes and 182 signal pathways to play the role of treating BA and AC. There were 131 active compounds related to key targets. The results of molecular docking showed that the important active compounds in SLBZP had good binding ability with the key targets. The key target-miRNA network showed that 94 miRNAs were predicted. Conclusion SLBZP has played the role of treating different diseases with the same treatment on BA and AC through the characteristics of multicompound, multitarget, and multipathway of traditional Chinese medicine, which provides a theoretical basis for explaining the mechanism and clinical application of SLBZP treating different diseases with the same treatment in BA and AC.
Collapse
|
23
|
Yu F, Li W, Wang L, Que S, Lu L. Characterization of grass carp FosB, Fosl2, JunD transcription factors in response to GCRV infection. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2020.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Díez-Tercero L, Delgado LM, Perez RA. Modulation of Macrophage Response by Copper and Magnesium Ions in Combination with Low Concentrations of Dexamethasone. Biomedicines 2022; 10:biomedicines10040764. [PMID: 35453514 PMCID: PMC9030383 DOI: 10.3390/biomedicines10040764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Macrophages have been deemed crucial for correct tissue regeneration, which is a complex process with multiple overlapping phases, including inflammation. Previous studies have suggested that divalent ions are promising cues that can induce an anti-inflammatory response, since they are stable cues that can be released from biomaterials. However, their immunomodulatory potential is limited in a pro-inflammatory environment. Therefore, we investigated whether copper and magnesium ions combined with low concentrations of the anti-inflammatory drug, dexamethasone (dex), could have a synergistic effect in macrophage, with or without pro-inflammatory stimulus, in terms of morphology, metabolic activity and gene expression. Our results showed that the combination of copper and dex strongly decreased the expression of pro-inflammatory markers, while the combination with magnesium upregulated the expression of IL-10. Moreover, in the presence of a pro-inflammatory stimulus, the combination of copper and dex induced a strong TNF-α response, suggesting an impairment of the anti-inflammatory actions of dex. The combination of magnesium and dex in the presence of a pro-inflammatory stimulus did not promote any improvement in comparison to dex alone. The results obtained in this study could be relevant for tissue engineering applications and in the design of platforms with a dual release of divalent ions and small molecules.
Collapse
Affiliation(s)
- Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - Luis M. Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Correspondence: (L.M.D.); (R.A.P.); Tel.: +34-935042000 (L.M.D. & R.A.P.)
| | - Roman A. Perez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Correspondence: (L.M.D.); (R.A.P.); Tel.: +34-935042000 (L.M.D. & R.A.P.)
| |
Collapse
|
25
|
Hwang D, Ryu HW, Park JW, Kim JH, Kim DY, Oh JH, Kwon OK, Han SB, Ahn KS. Effects of 3'-isovaleryl-4'-senecioylkhellactone from Peucedanum japonicum Thunberg on PMA-Stimulated Inflammatory Response in A549 Human Lung Epithelial Cells. J Microbiol Biotechnol 2022; 32:81-90. [PMID: 34818666 PMCID: PMC9628833 DOI: 10.4014/jmb.2107.07001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
Peucedanum japonicum Thunberg (PJT) has been used in traditional medicine to treat colds, coughs, fevers, and other inflammatory diseases. The goal of this study was to investigate whether 3'-isovaleryl-4'-senecioylkhellactone (IVSK) from PJT has anti-inflammatory effects on lung epithelial cells. The anti-inflammatory effects of IVSK were evaluated using phorbol 12-myristate 13-acetate (PMA)-stimulated A549 cells and regular human lung epithelial cells as a reference. IVSK reduced the secretion of the inflammatory mediators interleukin (IL)-8 and monocyte chemoattractant protein-1 (MCP-1), and the mRNA expression of IL-6, IL-8, MCP-1, and IL-1β. Additionally, it inhibited the phosphorylation of IκB kinase (IKK), p65, Iκ-Bα, and mitogen-activated protein kinases (MAPKs) p38, JNK, and ERK in A549 cells stimulated with PMA. Moreover, the binding affinity of activator protein-1 (AP-1) and nuclear factor-κB (NF-κB) was significantly reduced in the luciferase assay, while nuclear translocation was markedly inhibited by IVSK in the immunocytochemistry. These findings indicate that IVSK can protect against inflammation through the AP-1 and NF-κB pathway and could possibly be used as a lead compound for the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Daseul Hwang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea,College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jung-Hee Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Doo-Young Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jae-Hoon Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea,College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea,
O.K. Kwon E-mail:
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea,Corresponding authors S.B. Han E-mail:
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea,
K.S. Ahn E-mail: Phone: +82-43-240-6134 Fax: +82-43-240-6129
| |
Collapse
|
26
|
Shivaleela B, Srushti SC, Shreedevi SJ, Babu RL. Thalidomide-based inhibitor for TNF-α: designing and Insilico evaluation. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-021-00393-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Inflammatory diseases are the vast array of disorders caused by inflammation. During most inflammatory events, many cytokines expressions were modulated, and one such cytokine is tumor necrosis factor-alpha (TNF-α). TNF-α is mainly secreted by monocytes and macrophages. Notably, it has been proposed as a therapeutic target for several diseases. The anti-TNF biology approach is mainly based on monoclonal antibodies. The fusion protein and biosimilars are prevalent in treating inflammation for decades. Only a few small molecule inhibitors are available to inhibit the expression of TNF-α, and one such promising drug was thalidomide. Therefore, the study was carried out to design thalidomide-based small molecule inhibitors for TNF-α. The main objective of our study is to design thalidomide analogs to inhibit TNF-α using the insilico approach.
Results
Several thalidomide analogs were designed using chemsketch. After filtration of compounds through ‘Lipinski rule of 5’ by Molinspiration tool, as a result, five compounds were selected. All these compounds were subjected to molecular docking, and the study showed that all five compounds had good binding energy. However, based on ADMET predictions, two compounds (S3 and S5) were eliminated.
Conclusions
Our preliminary results suggest that S1, S2, S4 compounds showed potential ligand binding capacity with TNF-α and, interestingly, with limited or no toxicity. Our preliminary investigation and obtained results have fashioned more interest for further in vitro studies.
Collapse
|
27
|
Wang X, Zhang Y, Wuyun K, Gong H. Therapeutic effect and mechanism of 4‑phenyl butyric acid on renal ischemia‑reperfusion injury in mice. Exp Ther Med 2021; 23:144. [PMID: 35069825 PMCID: PMC8756420 DOI: 10.3892/etm.2021.11067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 08/06/2021] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the effects and possible mechanism of 4-phenylbutyric acid (4-PBA) on renal ischemia-reperfusion injury (RIRI) in mice. A RIRI model of HK-2 cells was constructed using hypoxia/reoxygenation (H/R) treatment. Dexmedetomidine and 4-PBA were used to treat the cells before and after modeling. Apoptosis and expression levels of cyclophilin D (CypD), cytochrome c, eukaryotic translation initiation factor 2α (eIF2α), glucose-regulated protein 78 (GRP78), intercellular adhesion molecule (ICAM)-1 and vascular adhesion molecule (VCAM)-1 were measured using flow cytometry, western blotting and immunohistochemistry. The renal volume, weight and renal arterial resistance index (RRI) were determined using the renal ischemia model. Compared with untreated model cells, 4-PBA treatment significantly decreased apoptosis and the expression levels of CypD, Cytochrome c, eIF2α and GRP78 in HK-2 cells. There was no significant change in renal volume and weight after modeling, but RRI was significantly decreased after 4-PBA treatments in the model. Western blotting and immunohistochemistry analysis demonstrated that 4-PBA treatment also significantly decreased the expression of ICAM-1 and VCAM-1. Overall, 4-PBA had a therapeutic effect on RIRI in mice. This protection may be mediated by decreasing the expression levels of CypD, Cytochrome c, eIF2α and GRP78, and subsequent reduction of cellular oxygen free radicals and apoptosis, leading to an alleviated endoplasmic reticulum stress response and RIRI.
Collapse
Affiliation(s)
- Xinlei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Kun Wuyun
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haixia Gong
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
28
|
Fan HC, Chang FW, Pan YR, Yu SI, Chang KH, Chen CM, Liu CA. Approach to the Connection between Meconium Consistency and Adverse Neonatal Outcomes: A Retrospective Clinical Review and Prospective In Vitro Study. CHILDREN (BASEL, SWITZERLAND) 2021; 8:1082. [PMID: 34943278 PMCID: PMC8700184 DOI: 10.3390/children8121082] [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: 09/15/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Whether meconium-stained amniotic fluid (MSAF) serves as an indicator of fetal distress is under debate; however, the presence of MSAF concerns both obstetricians and pediatricians because meconium aspiration is a major contributor to neonatal morbidity and mortality, even with appropriate treatment. The present study suggested that thick meconium in infants might be associated with poor outcomes compared with thin meconium based on chart reviews. In addition, cell survival assays following the incubation of various meconium concentrations with monolayers of human epithelial and embryonic lung fibroblast cell lines were consistent with the results obtained from chart reviews. Exposure to meconium resulted in the significant release of nitrite from A549 and HEL299 cells. Medicinal agents, including dexamethasone, L-Nω-nitro-arginine methylester (L-NAME), and NS-398 significantly reduced the meconium-induced release of nitrite. These results support the hypothesis that thick meconium is a risk factor for neonates who require resuscitation, and inflammation appears to serve as the primary mechanism for meconium-associated lung injury. A better understanding of the relationship between nitrite and inflammation could result in the development of promising treatments for meconium aspiration syndrome (MAS).
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan;
- Department of Medica research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan; (Y.-R.P.); (S.-I.Y.); (K.-H.C.)
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan;
| | - Fung-Wei Chang
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Ying-Ru Pan
- Department of Medica research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan; (Y.-R.P.); (S.-I.Y.); (K.-H.C.)
| | - Szu-I Yu
- Department of Medica research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan; (Y.-R.P.); (S.-I.Y.); (K.-H.C.)
| | - Kuang-Hsi Chang
- Department of Medica research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan; (Y.-R.P.); (S.-I.Y.); (K.-H.C.)
| | - Chuan-Mu Chen
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan;
- The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ching-Ann Liu
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
- Neuroscience Center, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
| |
Collapse
|
29
|
Ilex paraguariensis extract as an alternative to pain medications. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2021; 71:383-398. [PMID: 36654097 DOI: 10.2478/acph-2021-0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 01/20/2023]
Abstract
Pain is a common and distressing symptom of many diseases and its clinical treatment generally involves analgesics and anti-inflammatory drugs. This study evaluated the toxicity of Ilex paraguariensis A. St.-Hil. (Aquifoliaceae) aqueous extract (leaves, petioles and branches) and its performance in a nociceptive response. Hepatotoxicity, psycho-stimulant test and evaluation of enzyme markers for liver damage were also tested. Chromatographic analysis by UPLC-MS demonstrated a series of isomeric monocaffeoylquinic acids, isomers of dicaffeoylquinic acid, flavonol glycosides, and saponins. Phase I and II of nociception were obtained for meloxicam, dexamethasone and aqueous Ilex paraguariensis extract. Ilex paraguariensis extract concentration was negatively correlated (R = -0.887) with alanine aminotransferase (p < 0.05) in acetaminophen-induced hepatotoxicity test, indicating hepatoprotective activity of this extract. Ilex paraguariensis extract also presented analgesic properties equivalent to drugs that already have proven efficacy. Notably, the administration of multiple doses of Ilex paraguariensis extract was considered safe from the therapeutic point of view.
Collapse
|
30
|
Ferreira Junior NC, dos Santos Pereira M, Francis N, Ramirez P, Martorell P, González-Lizarraga F, Figadère B, Chehin R, Del Bel E, Raisman-Vozari R, Michel PP. The Chemically-Modified Tetracycline COL-3 and Its Parent Compound Doxycycline Prevent Microglial Inflammatory Responses by Reducing Glucose-Mediated Oxidative Stress. Cells 2021; 10:cells10082163. [PMID: 34440932 PMCID: PMC8392055 DOI: 10.3390/cells10082163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
We used mouse microglial cells in culture activated by lipopolysaccharide (LPS) or α-synuclein amyloid aggregates (αSa) to study the anti-inflammatory effects of COL-3, a tetracycline derivative without antimicrobial activity. Under LPS or αSa stimulation, COL-3 (10, 20 µM) efficiently repressed the induction of the microglial activation marker protein Iba-1 and the stimulated-release of the pro-inflammatory cytokine TNF-α. COL-3′s inhibitory effects on TNF-α were reproduced by the tetracycline antibiotic doxycycline (DOX; 50 µM), the glucocorticoid dexamethasone, and apocynin (APO), an inhibitor of the superoxide-producing enzyme NADPH oxidase. This last observation suggested that COL-3 and DOX might also operate themselves by restraining oxidative stress-mediated signaling events. Quantitative measurement of intracellular reactive oxygen species (ROS) levels revealed that COL-3 and DOX were indeed as effective as APO in reducing oxidative stress and TNF-α release in activated microglia. ROS inhibition with COL-3 or DOX occurred together with a reduction of microglial glucose accumulation and NADPH synthesis. This suggested that COL-3 and DOX might reduce microglial oxidative burst activity by limiting the glucose-dependent synthesis of NADPH, the requisite substrate for NADPH oxidase. Coherent with this possibility, the glycolysis inhibitor 2-deoxy-D-glucose reproduced the immunosuppressive action of COL-3 and DOX in activated microglia. Overall, we propose that COL-3 and its parent compound DOX exert anti-inflammatory effects in microglial cells by inhibiting glucose-dependent ROS production. These effects might be strengthened by the intrinsic antioxidant properties of DOX and COL-3 in a self-reinforcing manner.
Collapse
Affiliation(s)
- Nilson Carlos Ferreira Junior
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Maurício dos Santos Pereira
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Nour Francis
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paola Ramirez
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paula Martorell
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Florencia González-Lizarraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| | - Rosana Chehin
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| |
Collapse
|
31
|
Zhang Y, Yu Y, Li G, Zhang X, Wu Z, Lin L. Bioadhesive glycosylated nanoformulations for extended trans-corneal drug delivery to suppress corneal neovascularization. J Mater Chem B 2021; 9:4190-4200. [PMID: 33997882 DOI: 10.1039/d1tb00229e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Eye-drop formulations as conventional regimens to tackle ocular diseases are far from efficient due to the rapid clearance by eye tears and the blockage of the corneal epithelium barrier. Here, we describe a bioadhesive glycosylated nanoplatform with boric acid pendants as a drug carrier for noninvasive trans-corneal delivery of drugs to treat corneal neovascularization (CNV), a serious corneal disease resulting in significant vision impairment. This biocompatible nanoplatform is formulated from a synthetic amphiphilic boric acid-based copolymer self-assembling to form highly stable micelles with a high loading capacity for dexamethasone (DEX). The nanoplatform is demonstrated to be in contact with the corneal epithelium for a long period under the bioadhesive function of boric acid modules and releases the drug over 96 h in a controlled manner. Our results also suggest that the nanoplatform can be efficiently internalized by corneal epithelial cells in vitro and realize transcytosis in vivo to greatly enhance the transcorneal penetration of the loaded drugs into the pathological corneal stroma. On topical application against rat corneal alkali burn, the nanoformulation presents more robust efficacy on neovascularization suppression and inflammation elimination than free DEX with a negligible effect on normal tissues. This bioadhesive strategy which focuses on extending ocular drug retention and improving trans-corneal drug delivery not only highlights an approach for alternative noninvasive therapy of CNV but also provides a versatile paradigm for other biomedical applications by overcoming protective barriers.
Collapse
Affiliation(s)
- Yanlong Zhang
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China. and Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China and Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yunjian Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Gang Li
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China. and Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| | - Ling Lin
- State Key Laboratory of Precision Measurement Technology and Instrument, School of Precision Instruments & Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China. and Tianjin Key Laboratory of Biomedical Detection Techniques & Instruments, Tianjin University, Tianjin 300072, China
| |
Collapse
|
32
|
Aksoy F, Ege B, Tosun S. The effect of pre-operative submucosal administration of dexamethasone, tramadol, articaine on the success rate of inferior alveolar nerve block on mandibular molars with symptomatic irreversible pulpitis: A randomized, double-blind placebo-controlled clinical trial. Int Endod J 2021; 54:1982-1992. [PMID: 34347895 DOI: 10.1111/iej.13604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022]
Abstract
AIM To establish the effects of submucosal tramadol, dexamethasone and articaine on the success of inferior alveolar nerve blocks (IANB) during root canal treatment of mandibular molars with symptomatic irreversible pulpitis (SIP). METHODOLOGY In this randomized double-blind, controlled clinical trial, 120 patients with the diagnosis of SIP in their mandibular first or second molars were included and randomly divided into four groups (n = 30). The control group received normal saline and three experimental groups received a single dose of dexamethasone (8 mg/2 mL), or tramadol (100 mg/2 mL) or articaine (4% / 2 mL). The pre-operative pain levels of the patients were measured with the Heft-Parker visual analogue scale (HP VAS). All patients received standard IANB of 4% articaine with 1:200000 epinephrine. Following the observation of lip numbness, submucosal injections were administered into the mucobuccal fold adjacent to the mandibular molars. After 15 min, standard root canal treatment was initiated, and the pain intensity levels recorded from the access cavity preparation to pulp extirpation were measured with HP VAS. The duration of the anaesthesia was also evaluated. The experimental groups were compared using one-way ANOVA or Kruskal-Wallis H-test. The groups that were significantly different were compared pairwise using the Tukey Multiple Comparison test. The Chi-square test was used to compare the categorical variables. RESULTS The submucosal administration of articaine significantly increased the success rate to 63% in comparison with the control group that received submucosal saline (p < .05). The success rate of pulpal anaesthesia was 37% in the control group, 57% in the tramadol group and 47% in the dexamethasone group, with no significant difference in the success rate among these groups. In the dexamethasone group, the duration of the anaesthetic effect of IANB was significantly longer than those in the other groups (p < .001). CONCLUSIONS In patients with SIP, pre-operative submucosal administration of articaine increased the success rate of IANB, while administration of dexamethasone increased the duration of anaesthesia. These agents can be used in cases where effective anaesthesia cannot be obtained during root canal treatments.
Collapse
Affiliation(s)
- Fatih Aksoy
- Department of Endodontics, Faculty of Dentistry, Adıyaman University, Adıyaman, Turkey
| | - Bilal Ege
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Adıyaman University, Adıyaman, Turkey
| | - Samet Tosun
- Department of Endodontics, Faculty of Dentistry, Pamukkale University, Denizli, Turkey
| |
Collapse
|
33
|
Chang WT, Hong MY, Chen CL, Hwang CY, Tsai CC, Chuang CC. Mutant glucocorticoid receptor binding elements on the interleukin-6 promoter regulate dexamethasone effects. BMC Immunol 2021; 22:24. [PMID: 33771121 PMCID: PMC7995394 DOI: 10.1186/s12865-021-00413-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/14/2021] [Indexed: 12/15/2022] Open
Abstract
Background Glucocorticoids (GCs) have been extensively used as essential modulators in clinical infectious and inflammatory diseases. The GC receptor (GR) is a transcription factor belonging to the nuclear receptor family that regulates anti-inflammatory processes and releases pro-inflammatory cytokines, such as interleukin (IL)-6. Results Five putative GR binding sites and other transcriptional factor binding sites were identified on theIL-6 promoter, and dexamethasone (DEX) was noted to reduce the lipopolysaccharide (LPS)-induced IL-6 production. Among mutant transcriptional factor binding sites, nuclear factor-kappa B (NF-κB), activator protein (AP)-1, and specificity protein (Sp)1–2 sites reduced basal and LPS-induced IL-6 promoter activities through various responses. The second GR binding site (GR2) was noted to play a crucial role in both basal and inducible promoter activities in LPS-induced inflammation. Conclusions We concluded that selective GR2 modulator might exert agonistic and antagonistic effects and could activate crucial signaling pathways during the LPS-stimulated inflammatory process. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00413-z.
Collapse
Affiliation(s)
- Wen-Teng Chang
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, 701, Taiwan
| | - Ming-Yuan Hong
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Liang Chen
- Department of Physical Therapy, I-Shou University, Kaohsiung, Taiwan
| | - Chi-Yuan Hwang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan, 701, Taiwan
| | - Chia-Chang Chuang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
34
|
Caputo LS, Campos MIC, Dias HJ, Crotti AEM, Fajardo JB, Vanelli CP, Presto ÁCD, Alves MS, Aarestrup FM, Paula ACC, Da Silva Filho AA, Aarestrup BJV, Pereira OS, Corrêa JODA. Copaiba oil suppresses inflammation in asthmatic lungs of BALB/c mice induced with ovalbumin. Int Immunopharmacol 2020; 80:106177. [PMID: 32007706 DOI: 10.1016/j.intimp.2019.106177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/05/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022]
Abstract
Asthma is a chronic inflammatory disease that represents high hospitalizations and deaths in world. Copaiba oil (CO) is popularly used for relieving asthma symptoms and has already been shown to be effective in many inflammation models. This study aimed to investigate the immunomodulatory relationship of CO in ovalbumin (OVA)-induced allergic asthma. The composition of CO sample analyzed by GC and GC-MS and the toxicity test was performed in mice at doses of 50 or 100 mg/kg (by gavage). After, the experimental model of allergic asthma was induced with OVA and mice were orally treated with CO in two pre-established doses. The inflammatory infiltrate was evaluated in bronchoalveolar lavage fluid (BALF), while cytokines (IL-4, IL-5, IL-17, IFN-γ, TNF-α), IgE antibody and nitric oxide (NO) production was evaluated in BALF and lung homogenate (LH) of mice, together with the histology and histomorphometry of the lung tissue. CO significantly attenuated the number of inflammatory cells in BALF, suppressing NO production and reducing the response mediated by TH2 and TH17 (T helper) cells in both BALF and LH. Histopathological and histomorphometric analysis confirmed that CO significantly reduced the numbers of inflammatory infiltrate in the lung tissue, including in the parenchyma area. Our results indicate that CO has an effective in vivo antiasthmatic effect.
Collapse
Affiliation(s)
- Ludmila S Caputo
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Maria Inês C Campos
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Herbert J Dias
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Av. Bandeirantesn° 3900, 14040-901 Ribeirão Preto, SP, Brazil
| | - Antônio E M Crotti
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Av. Bandeirantesn° 3900, 14040-901 Ribeirão Preto, SP, Brazil
| | - Júlia B Fajardo
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Chislene P Vanelli
- Health Department, Faculty of Medical Sciences and Health of Juiz de Fora (SUPREMA), Alameda Salvaterra n° 200, Salvaterra, 36.033-003 Juiz de Fora, MG, Brazil
| | - Álvaro C D Presto
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Maria S Alves
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Fernando M Aarestrup
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Ana Claudia C Paula
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Ademar A Da Silva Filho
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Beatriz J V Aarestrup
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Olavo S Pereira
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - José Otávio do A Corrêa
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil.
| |
Collapse
|
35
|
Therapeutic Efficacy of Intratendinous Delivery of Dexamethasone Using Porous Microspheres for Amelioration of Inflammation and Tendon Degeneration on Achilles Tendinitis in Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5052028. [PMID: 32090096 PMCID: PMC6996678 DOI: 10.1155/2020/5052028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/24/2019] [Indexed: 11/17/2022]
Abstract
Achilles tendinitis caused by overuse, aging, or gradual wear induces pain, swelling, and stiffness of Achilles tendon and leads to tendon rupture. This study was performed to investigate the suppression of inflammation responses in interleukin-1β- (IL-1β-) stimulated tenocytes in vitro and the suppression of the progression of Achilles tendinitis-induced rat models in vivo using dexamethasone-containing porous microspheres (DEX/PMSs) for a sustained intratendinous DEX delivery. DEX from DEX/PMSs showed the sustained release of DEX. Treatment of IL-1β-stimulated tenocytes with DEX/PMSs suppressed the mRNA levels for COX-2, IL-1β, IL-6, and TNF-α. The intratendinous injection of DEX/PMSs into Achilles tendinitis rats both decreased the mRNA levels for these cytokines and increased mRNA levels for anti-inflammatory cytokines IL-4 and IL-10 in tendon tissues. Furthermore, DEX/PMSs effectively prevented tendon degeneration by enhancing the collagen content and biomechanical properties. Our findings suggest that DEX/PMSs show great potential as a sustained intratendinous delivery system for ameliorating inflammation responses as well as tendon degeneration in Achilles tendinitis.
Collapse
|
36
|
Ai F, Zhao G, Lv W, Liu B, Lin J. Dexamethasone induces aberrant macrophage immune function and apoptosis. Oncol Rep 2019; 43:427-436. [PMID: 31894280 PMCID: PMC6967116 DOI: 10.3892/or.2019.7434] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/22/2019] [Indexed: 12/28/2022] Open
Abstract
Glucocorticoids (GCs) are known potent clinical drugs, however, their mode of action is still complex and debatable. Macrophages are the most important target of GCs and play a key role in tumor immunity in vivo, but their relationship is also controversial. In the present study, the lentivirus system was used to overexpress and knock down the level of transcription factor Krüppel-like factor 9 (KLF9). The results revealed that dexamethasone (Dex) induced ROS generation and mitochondria-dependent apoptosis in RAW 264.7 cells via the KLF9. In addition, overexpression of KLF9 significantly increased apoptosis of RAW 264.7 cells. Notably, ELISA assay revealed that increased expression of KLF9 inhibited LPS-induced COX-2 expression and reduced COX-2-derived prostaglandin E2 and pro-inflammatory cytokine secretion. Furthermore, a co-culture system was used to reveal that overexpression of KLF9 in RAW 264.7 cells promoted HepG2 cell survival. In summary, it is reported that KLF9 promoted apoptosis of proinflammatory macrophages, and suppressed the antitumor effects, which can be selectively targeted by GCs as a novel mechanism to suppress antineoplastic activity.
Collapse
Affiliation(s)
- Fulu Ai
- Department of General Surgery (VIP ward), Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Guohua Zhao
- Department of General Surgery (VIP ward), Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Wu Lv
- Department of General Surgery (VIP ward), Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Bin Liu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Jie Lin
- Department of General Surgery (VIP ward), Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
37
|
Guan M, Ma H, Fan X, Chen X, Miao M, Wu H. Dexamethasone alleviate allergic airway inflammation in mice by inhibiting the activation of NLRP3 inflammasome. Int Immunopharmacol 2019; 78:106017. [PMID: 31780368 DOI: 10.1016/j.intimp.2019.106017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/11/2019] [Accepted: 10/29/2019] [Indexed: 01/20/2023]
Abstract
Dexamethasone (DEX) is the mainstay treatment for asthma, which is a common chronic airway inflammation disease. However, the mechanism of DEX resolute symptoms of asthma is not completely clear. Here, we aimed to analyze the effect of DEX on airway inflammation in OVA-induced mice and whether this effect is related to the inhibition of the activation of NLRP3 inflammasome. Female (C57BL/6) mice were used to establish the allergic airway inflammation model by inhalation OVA. The number of inflammatory cells in the bronchi alveolar lavage fluid (BALF) was counted by Swiss-Giemsa staining, and the contents of IL-1β, IL-18, IL-5 and IL-17 were detected by ELISA. The degree of inflammatory cells infiltration and mucous cells proliferation in lung tissue were separately observed by H&E and PAS staining. The proteins expression of NLRP3, pro-caspase-1, caspase-1, IL-1β, IL-6 and IL-17 in lung tissue were detected by Western blotting. We found that DEX significantly inhibited OVA-induced inflammatory cells infiltration, airway mucus secretion and goblet cell proliferation in mice. The total and classified numbers of inflammatory cells and the levels of IL-1β, IL-18, IL-5 and IL-17 in the BALF of the experimental group were significantly lower than those of the model group after DEX treatment. DEX also significantly inhibited the activity of NLRP3 inflammasome and reduced the protein contents of Pro-Caspase-1, Caspase-1, Capase-1/Pro-Caspase-1, IL-1β, IL-6 and IL-17 in lung tissues. Our study suggested that DEX alleviates allergic airway inflammation by inhibiting the activity of NLRP3 inflammasome and the levels of IL-1β and IL-18.
Collapse
Affiliation(s)
- Minglong Guan
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Hengli Ma
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| | - Xu Chen
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Manli Miao
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Institute of Respiratory Diseases, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| | - Huimei Wu
- Department of Geriatric Respiratory and Critical Care, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Geriatric Institute, Jixi Road 218, Hefei, Anhui 230022, PR China; Anhui Key Laboratory of Geriatric Molecular Medicine, Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China
| |
Collapse
|
38
|
Zhang W, Li X, Zhang Y. Rho-kinase inhibitor attenuates airway mucus hypersecretion and inflammation partly by downregulation of IL-13 and the JNK1/2-AP1 signaling pathway. Biochem Biophys Res Commun 2019; 516:571-577. [PMID: 31235256 DOI: 10.1016/j.bbrc.2019.06.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 11/26/2022]
Abstract
We measured the effect of Rho-kinase on inflammation and mucus hypersecretion in the airways of mouse models of asthma. Additionally, we aimed to determine if these effects were the result of JNK 1/2-AP1 pathway inhibition.We sensitized and challenged female C57BL/6 mice using house dust mites (HDM) followed by treatment with an inhibitor of Rho-kinase. Lung tissue was harvested to evaluate inflammation and mucus secretion in the airways of asthma mice. Cytokine expression in broncho-alveolar lavage fluid (BALF) was established by ELISA and airway responsiveness, and was determined by the invasive lung function test. JNK1/2, p-JNK1/2, AP-1, and p-AP-1 protein expression was determined by Western blot analysis. Asthma model mice that were treated with Rho-kinase inhibitor showed a significantly decrease in inflammation score, inflammatory cells, and airway responsiveness. Additionally, we found that IL-13 expressions in BALF and mucus secretion were decreased in HDM-challenged mice treated with Rho-kinase inhibitor. Furthermore, Rho-kinase inhibitor treatment decreased the expression of JNK1/2 and AP-1 phosphorylation. Our findings indicated that the Rho-kinase inhibitor decreased HDM-induced mucus secretion as well as airway inflammation in asthma mice through regulation of the JNK1/2-AP-1 pathway.
Collapse
Affiliation(s)
- Wenqin Zhang
- Department of geriatric medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xin Li
- Department of Respiratory Medicine, Tianjin Medical University General Hospital, Tianjin, 300073, China
| | - Yun Zhang
- Department of geriatric medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
39
|
Silwedel C, Fehrholz M, Speer CP, Ruf KC, Manig S, Glaser K. Differential modulation of pulmonary caspases: Is this the key to Ureaplasma-driven chronic inflammation? PLoS One 2019; 14:e0216569. [PMID: 31067276 PMCID: PMC6506144 DOI: 10.1371/journal.pone.0216569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/23/2019] [Indexed: 12/26/2022] Open
Abstract
Although accepted agents in chorioamnionitis and preterm birth, the role of Ureaplasma species (spp.) in inflammation-driven morbidities of prematurity, including the development of bronchopulmonary dysplasia, remains controversial. To add to scarce in vitro data addressing the pro-inflammatory capacity of Ureaplasma spp., pulmonary epithelial-like A549 cells and human pulmonary microvascular endothelial cells (HPMEC) were incubated with Ureaplasma (U.) urealyticum, U. parvum, and Escherichia coli lipopolysaccharide (LPS). Ureaplasma isolates down-regulated caspase mRNA levels in A549 cells (caspase 8: p<0.001, 9: p<0.001, vs. broth), while increasing caspase protein expression, enzyme activity, and cell death in HPMEC (active caspase 3: p<0.05, caspase 8: p<0.05, active caspase 9: p<0.05, viability: p<0.05). LPS, contrarily, induced caspase mRNA expression in HPMEC (caspase 3: p<0.01, 4: p<0.001, 5: p<0.001, 8: p<0.001, vs. control), but not in A549 cells, and did not affect enzyme activity or protein levels in either cell line. LPS, but neither Ureaplasma isolate, enhanced mRNA expression of pro-inflammatory interleukin (IL)-6 in both A549 (p<0.05, vs. control) and HPMEC (p<0.001) as well as tumor necrosis factor-α (p<0.01), IL-1β (p<0.001), and IL-8 (p<0.05) in HPMEC. We are therefore the first to demonstrate a differential modulation of pulmonary caspases by Ureaplasma spp. in vitro. Ureaplasma-driven enhanced protein expression and activity of caspases in pulmonary endothelial cells result in cell death and may cause structural damage. Down-regulated caspase mRNA in pulmonary epithelial cells, contrarily, may indicate Ureaplasma-induced inhibition of apoptosis and prevent effective immune responses. Both may ultimately contribute to chronic Ureaplasma colonization and long-term pulmonary inflammation.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Markus Fehrholz
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Christian P. Speer
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Katharina C. Ruf
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Steffi Manig
- Institute of Hygiene and Microbiology, University of Wuerzburg, Wuerzburg, Germany
| | - Kirsten Glaser
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
40
|
Eshraghi AA, Jung HD, Mittal R. Recent Advancements in Gene and Stem Cell-Based Treatment Modalities: Potential Implications in Noise-Induced Hearing Loss. Anat Rec (Hoboken) 2019; 303:516-526. [PMID: 30859735 DOI: 10.1002/ar.24107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/24/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022]
Abstract
Noise-induced hearing loss (NIHL) poses a significant burden on not only the economics of health care but also the quality of life of an individual, as we approach an unprecedented age of longevity. In this article, we will delineate the current landscape of management of NIHL. We discuss the most recent results from in vitro and in vivo studies that determine the effectiveness of established pharmacotherapy such as corticosteroid and potential emerging therapies like N-acetyl cysteine and neurotrophins (NTs), as well as highlight ongoing clinical trials for these therapeutic agents. We present an overview of how the recent advancements in the field of gene-based and stem cell-based therapies can help in developing effective therapeutic strategies for NIHL. Gene-based therapies have shown exciting results demonstrating cochlear cellular regeneration using Atoh1, NRF2 as well as NT gene therapy employing viral vectors. In addition, we will discuss the recent advancements in genome-editing technologies, such as CRISPR/Cas9, and its potential role in NIHL therapy. We will further discuss the current state of stem cell therapy as it pertains to treating neurodegenerative conditions including NIHL. Embryonic stem cells, adult-derived stem cells, and induced pluripotent stem cells all represent an enticing reservoir of replacing damaged cells as a result of NIHL. Finally, we will discuss the barriers that need to be overcome to translate these promising treatment modalities to the clinical practice in pursuit of improving quality of life of patients having NIHL. Anat Rec, 303:516-526, 2020. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Adrien A Eshraghi
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Hyunseo D Jung
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| | - Rahul Mittal
- Department of Otolaryngology, Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
41
|
Choi BS, Kim YJ, Choi JS, Lee HJ, Lee CJ. Obtusifolin isolated from the seeds of Cassia obtusifolia regulates the gene expression and production of MUC5AC mucin in airway epithelial cells via affecting NF-κB pathway. Phytother Res 2019; 33:919-928. [PMID: 30632219 DOI: 10.1002/ptr.6284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/08/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
We investigated whether obtusin, obtusifolin, and cassiaside isolated from the seeds of Cassia obtusifolia inhibit the gene expression and production of airway mucin 5AC (MUC5AC). Confluent NCI-H292 cells were pretreated with obtusin, obtusifolin, or cassiaside for 30 min and then stimulated with epidermal growth factor (EGF), phorbol 12-myristate 13-acetate (PMA), or tumor necrosis factor-α (TNF-α) for 24 hr. The MUC5AC mucin gene expression was measured by reverse transcription-polymerase chain reaction. Production of MUC5AC mucin protein was measured by enzyme-linked immunosorbent assay. To elucidate the action mechanism of obtusifolin, effect of obtusifolin on PMA-induced nuclear factor kappa B (NF-κB) signaling pathway was investigated by western blot analysis. Obtusin, obtusifolin, or cassiaside inhibited the expression of MUC5AC mucin gene and the production of MUC5AC mucin protein, induced by EGF, PMA, or TNF-α. Obtusifolin inhibited PMA-induced activation (phosphorylation) of inhibitory kappa B kinase, and thus phosphorylation and degradation of inhibitory kappa B alpha. Obtusifolin inhibited PMA-induced nuclear translocation of NF-κB p65. These results suggest that obtusifolin can regulate the production and gene expression of mucin by acting on airway epithelial cells through regulation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Byung-Soo Choi
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Yu-Jin Kim
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Jae Sue Choi
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
| | - Hyun Jae Lee
- Smith Liberal Arts College and Department of Addiction Science, Graduate School, Sahmyook University, Seoul, Korea
| | - Choong Jae Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan, Korea
| |
Collapse
|
42
|
Babu RL, Naveen Kumar M, Patil RH, Kiran Kumar KM, Devaraju KS, Ramesh GT, Sharma SC. Forskolin and Phorbol 12-myristate 13-acetate modulates the expression pattern of AP-1 factors and cell cycle regulators in estrogen-responsive MCF-7 cells. Genes Dis 2018; 6:159-166. [PMID: 31194000 PMCID: PMC6545452 DOI: 10.1016/j.gendis.2018.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Activator protein-1 (AP-1) transcription factor is a key component of many signal transduction pathways involved in the regulation of cellular processes and controls rapid responses of mammalian cells when exposed to the variety of stimulus. The phorbol 12-myristate 13-acetate and Forskolin (Fo) are well-known kinase activators/stimulators of Protein Kinase C (PKC) and Protein Kinase A (PKA) respectively. Importantly, these kinases are found to be present in transitional points of many cell signaling pathways, especially those involved in proliferation. The stimulating effect of PKC and PKA on the expression of AP-1 factors in MCF-7 breast cell proliferation is not well characterized. Hence, the role of PKC by PMA treatment and the role of PKA by using Fo in MCF-7 cells is investigated. Where, cells treated with PMA showed increased cell proliferation, while Fo had no effect, but inhibited the PMA induced proliferation. The RT-PCR results showed the PMA induced c-Jun, c-Fos and Fra-1 expressions compared to control and Fo. However, Fo in combination with PMA, inhibit the PMA induced above mRNA expressions where Fo alone has no effect. Western blot studies validated the c-Jun expressions in PMA treated MCF-7 cells. Further, PMA increases the mRNA expression of Cyclin-E1, Cyclin-D1, and CDK-4, whereas Fo decreases their expressions. Thus, mitogenic effect of PMA and inhibitory action of Fo on MCF-7 cells is probably enhanced via activation of AP-1 factors and concomitant action of cell cycle regulators in the downstream singling cascade.
Collapse
Affiliation(s)
- R L Babu
- Department of Bioinformatics and Biotechnology, Akkamahadevi Women's University, Jnanashakthi Campus, Vijayapura, 586 108, Karnataka, India.,Department of Biology, Center for Biotechnology and Biomedical Sciences, Norfolk State University, Norfolk, VA, USA
| | - M Naveen Kumar
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru, 560 056, Karnataka, India
| | - Rajeshwari H Patil
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru, 560 056, Karnataka, India
| | - K M Kiran Kumar
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru, 560 056, Karnataka, India
| | - K S Devaraju
- Department of Biochemistry, Karnatak University, Dharwad, 580003, Karnataka, India
| | - Govindarajan T Ramesh
- Department of Biology, Center for Biotechnology and Biomedical Sciences, Norfolk State University, Norfolk, VA, USA
| | - S Chidananda Sharma
- Department of Microbiology and Biotechnology, Bangalore University, Jnana Bharathi, Bengaluru, 560 056, Karnataka, India
| |
Collapse
|
43
|
The Role of BPIFA1 in Upper Airway Microbial Infections and Correlated Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2021890. [PMID: 30255091 PMCID: PMC6140130 DOI: 10.1155/2018/2021890] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/04/2018] [Accepted: 08/07/2018] [Indexed: 12/16/2022]
Abstract
The mucosa is part of the first line of immune defense against pathogen exposure in humans and prevents viral and bacterial infection of the soft palate, lungs, uvula, and nasal cavity that comprise the ear-nose-throat (ENT) region. Bactericidal/permeability-increasing fold containing family A, member 1 (BPIFA1) is a secretory protein found in human upper aerodigestive tract mucosa. This innate material is secreted in mucosal fluid or found in submucosal tissue in the human soft palate, lung, uvula, and nasal cavity. BPIFA1 is a critical component of the innate immune response that prevents upper airway diseases. This review will provide a brief introduction of the roles of BPIFA1 in the upper airway (with a focus on the nasal cavity, sinus, and middle ear), specifically its history, identification, distribution in various human tissues, function, and diagnostic value in various upper airway infectious diseases.
Collapse
|
44
|
Sakai S, Nishida A, Ohno M, Inatomi O, Bamba S, Sugimoto M, Kawahara M, Andoh A. Astaxanthin, a xanthophyll carotenoid, prevents development of dextran sulphate sodium-induced murine colitis. J Clin Biochem Nutr 2018; 64:66-72. [PMID: 30705514 PMCID: PMC6348411 DOI: 10.3164/jcbn.18-47] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/11/2018] [Indexed: 12/11/2022] Open
Abstract
Astaxanthin is a xanthophyll carotenoid, which possesses strong scavenging effect on reactive oxygen species. In this study, we examined the effect of astaxanthin on dextran sulfate sodium (DSS)-induced colitis in mice. Experimental colitis was induced by the oral administration of 4% w/v DSS in tap water in C57BL/6J mice. Astaxanthin was mixed with a normal rodent diet (0.02 or 0.04%). Astaxanthin significantly ameliorated DSS-induced body weight loss and reduced the disease activity index. The ameliorating effects was observed in a dose-dependent manner. Immunochemical analyses showed that astaxanthin markedly suppressed DSS-induced histological inflammatory changes (inflammatory cell infiltration, edematous changes and goblet cell depletion). Plasma levels of malondialdehyde and 8-hydroxy-2-deoxyguanosine were significantly reduced by the administration of 0.04% astaxanthin. Astaxanthin significantly suppressed the mucosal mRNA expression of IL-1β, IL-6, TNF-α, IL-36α and IL-36γ. Astaxanthin blocked the DSS-induced translocation of NF-κB p65 and AP-1 (c-Jun) into the nucleus of mucosal epithelial cells, and also suppressed DSS-induced mucosal activation of MAPKs (ERK1/2, p38 and JNK). In conclusion, astaxanthin prevented the development of DSS-induced colitis via the direct suppression of NF-κB, AP-1 and MAPK activation. These findings suggest that astaxanthin is a novel candidate as a therapeutic option for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Shigeki Sakai
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Masashi Ohno
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Osamu Inatomi
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Shigeki Bamba
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Mitsushige Sugimoto
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Masahiro Kawahara
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | - Akira Andoh
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| |
Collapse
|