1
|
Xu Z, Wu Y, Zhao X, Zhou H. Integrating nontargeted metabolomics and RNA sequencing of dexamethasone-treated and untreated asthmatic mice reveals changes of amino acids and aminoacyl-tRNA in group 2 innate lymphoid cells. Int J Biol Macromol 2024; 283:137630. [PMID: 39547613 DOI: 10.1016/j.ijbiomac.2024.137630] [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: 08/21/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bronchial asthma is the most common multifactorial and heterogeneous disease in childhood. The glucocorticoid dexamethasone is a classic treatment for asthma. Research indicates that group 2 innate lymphoid cells (ILC2s) are crucial to the pathogenesis of asthma. However, few studies have focused on ILC2s metabolism and transcription. This study aims to establish an ovalbumin (OVA)-induced asthma model and a dexamethasone-treated asthma model to explore the regulation of lung ILC2s at the genetic and metabolic levels during the progression and remission of asthma, utilizing single-cell metabolomics and transcriptomics approaches. The results showed that ILC2s regulated the metabolic pathways and transcriptional levels of amino acids (such as arginine, proline, and histidine) and linoleic acid, as well as the metabolic biomarkers of arginine, urocanic acid, and linoleic acid in asthma. Additionally, the cytokine pathways and NF-γB pathways have been altered at the genetic level. At the same time, we revealed that dexamethasone regulates ILC2s amino acid and aminoacyl tRNA metabolism, as well as related genes, thereby alleviating asthma symptoms. Furthermore, we identified the genes Eno3 and Tap1, which are significantly associated with asthma. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to verify the accuracy of the RNA sequencing results. This study, for the first time, revealed the mechanistic changes of ILC2s in the development and treatment of asthma using multiomics techniques, laying a foundation for targeted therapies in asthma.
Collapse
Affiliation(s)
- Zhiwei Xu
- Department of Pediatrics, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Yaling Wu
- Department of Pediatrics, Bengbu Medical University, Bengbu, Anhui 233030, China
| | - Xiaoman Zhao
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui 230088, China
| | - Haoquan Zhou
- Department of Pediatrics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
2
|
Sahu B, Nookala S, Floden AM, Ambhore NS, Sathish V, Klug MG, Combs CK. House dust mite-induced asthma exacerbates Alzheimer's disease changes in the brain of the App NL-G-F mouse model of disease. Brain Behav Immun 2024; 121:365-383. [PMID: 39084541 PMCID: PMC11442016 DOI: 10.1016/j.bbi.2024.07.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) plaques, neuroinflammation, and neuronal death. Besides aging, various comorbidities increase the risk of AD, including obesity, diabetes, and allergic asthma. Epidemiological studies have reported a 2.17-fold higher risk of dementia in asthmatic patients. However, the molecular mechanism(s) underlying this asthma-associated AD exacerbation is unknown. This study was designed to explore house dust mite (HDM)-induced asthma effects on AD-related brain changes using the AppNL-G-F transgenic mouse model of disease. Male and female 8-9 months old C57BL/6J wild type and AppNL-G-F mice were exposed to no treatment, saline sham, or HDM extract every alternate day for 16 weeks for comparison across genotypes and treatment. Mice were euthanized at the end of the experiment, and broncho-alveolar lavage fluid (BALF), blood, lungs, and brains were collected. BALF was used to quantify immune cell phenotype, cytokine levels, total protein content, lactate dehydrogenase (LDH) activity, and total IgE. Lungs were sectioned and stained with hematoxylin and eosin, Alcian blue, and Masson's trichrome. Serum levels of cytokines and soluble Aβ1-40/42 were quantified. Brains were sectioned and immunostained for Aβ, GFAP, CD68, and collagen IV. Finally, frozen hippocampi and temporal cortices were used to perform Aβ ELISAs and cytokine arrays, respectively. HDM exposure led to increased levels of inflammatory cells, cytokines, total protein content, LDH activity, and total IgE in the BALF, as well as increased pulmonary mucus and collagen staining in both sexes and genotypes. Levels of serum cytokines increased in all HDM-exposed groups. Serum from the AppNL-G-F HDM-induced asthma group also had significantly increased soluble Aβ1-42 levels in both sexes. In agreement with this peripheral change, hippocampi from asthma-induced male and female AppNL-G-F mice demonstrated elevated Aβ plaque load and increased soluble Aβ 1-40/42 and insoluble Aβ 1-40 levels. HDM exposure also increased astrogliosis and microgliosis in both sexes of AppNL-G-F mice, as indicated by GFAP and CD68 immunoreactivity, respectively. Additionally, HDM exposure elevated cortical levels of several cytokines in both sexes and genotypes. Finally, HDM-exposed groups also showed a disturbed blood-brain-barrier (BBB) integrity in the hippocampus of AppNL-G-F mice, as indicated by decreased collagen IV immunoreactivity. HDM exposure was responsible for an asthma-like condition in the lungs that exacerbated Aβ pathology, astrogliosis, microgliosis, and cytokine changes in the brains of male and female AppNL-G-F mice that correlated with reduced BBB integrity. Defining mechanisms of asthma effects on the brain may identify novel therapeutic targets for asthma and AD.
Collapse
Affiliation(s)
- Bijayani Sahu
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Suba Nookala
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA
| | - Nilesh S Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Marilyn G Klug
- Department of Population health, School of Medicine and Health Sciences, USA
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037, USA.
| |
Collapse
|
3
|
Thon P, Rahmel T, Ziehe D, Palmowski L, Marko B, Nowak H, Wolf A, Witowski A, Orlowski J, Ellger B, Wappler F, Schwier E, Henzler D, Köhler T, Zarbock A, Ehrentraut SF, Putensen C, Frey UH, Anft M, Babel N, Sitek B, Adamzik M, Bergmann L, Unterberg M, Koos B, Rump K. AQP3 and AQP9-Contrary Players in Sepsis? Int J Mol Sci 2024; 25:1209. [PMID: 38279209 PMCID: PMC10816878 DOI: 10.3390/ijms25021209] [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: 12/11/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
Sepsis involves an immunological systemic response to a microbial pathogenic insult, leading to a cascade of interconnected biochemical, cellular, and organ-organ interaction networks. Potential drug targets can depict aquaporins, as they are involved in immunological processes. In immune cells, AQP3 and AQP9 are of special interest. In this study, we tested the hypothesis that these aquaporins are expressed in the blood cells of septic patients and impact sepsis survival. Clinical data, routine laboratory parameters, and blood samples from septic patients were analyzed on day 1 and day 8 after sepsis diagnosis. AQP expression and cytokine serum concentrations were measured. AQP3 mRNA expression increased over the duration of sepsis and was correlated with lymphocyte count. High AQP3 expression was associated with increased survival. In contrast, AQP9 expression was not altered during sepsis and was correlated with neutrophil count, and low levels of AQP9 were associated with increased survival. Furthermore, AQP9 expression was an independent risk factor for sepsis lethality. In conclusion, AQP3 and AQP9 may play contrary roles in the pathophysiology of sepsis, and these results suggest that AQP9 may be a novel drug target in sepsis and, concurrently, a valuable biomarker of the disease.
Collapse
Affiliation(s)
- Patrick Thon
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Tim Rahmel
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Dominik Ziehe
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Lars Palmowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Britta Marko
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Hartmuth Nowak
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
- Center for Artificial Intelligence, Medical Informatics and Data Science, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Alexander Wolf
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Andrea Witowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Jennifer Orlowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Björn Ellger
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Klinikum Westfalen, 44309 Dortmund, Germany;
| | - Frank Wappler
- Department of Anesthesiology and Operative Intensive Care Medicine, University of Witten/Herdecke, Cologne Merheim Medical School, 51109 Cologne, Germany;
| | - Elke Schwier
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Dietrich Henzler
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Thomas Köhler
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Alexander Zarbock
- Klinik für Anästhesiologie, Operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, 48149 Münster, Germany;
| | - Stefan Felix Ehrentraut
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, 53127 Bonn, Germany; (S.F.E.); (C.P.)
| | - Christian Putensen
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, 53127 Bonn, Germany; (S.F.E.); (C.P.)
| | - Ulrich Hermann Frey
- Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, 44625 Herne, Germany;
| | - Moritz Anft
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Herne, Germany; (M.A.); (N.B.)
| | - Nina Babel
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Herne, Germany; (M.A.); (N.B.)
| | - Barbara Sitek
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Michael Adamzik
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Lars Bergmann
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Matthias Unterberg
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Björn Koos
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| | - Katharina Rump
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (P.T.); (T.R.); (D.Z.); (L.P.); (B.M.); (H.N.); (A.W.); (J.O.); (B.S.); (M.A.); (L.B.); (M.U.); (B.K.)
| |
Collapse
|
4
|
Pei C, Jia N, Wang Y, Zhao S, Shen Z, Shi S, Huang D, Wu Y, Wang X, Li S, He Y, Wang Z. Notoginsenoside R1 protects against hypobaric hypoxia-induced high-altitude pulmonary edema by inhibiting apoptosis via ERK1/2-P90rsk-BAD ignaling pathway. Eur J Pharmacol 2023; 959:176065. [PMID: 37775017 DOI: 10.1016/j.ejphar.2023.176065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/03/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
High-altitude pulmonary edema (HAPE) is a potentially fatal disease. Notoginsenoside R1 is a novel phytoestrogen with anti-inflammatory, antioxidant and anti-apoptosis properties. However, its effects and underlying mechanisms in the protection of hypobaric hypoxia-induced HAPE rats remains unclear. This study aimed to explore the protective effects and underlying mechanisms of Notoginsenoside R1 in hypobaric hypoxia-induced HAPE. We found that Notoginsenoside R1 alleviated the lung tissue injury, decreased lung wet/dry ratio, and reduced inflammation and oxidative stress. Additionally, Notoginsenoside R1 ameliorated the changes in arterial blood gas, decreased the total protein concentration in bronchoalveolar lavage fluid, and inhibited the occurrence of apoptosis caused by HAPE. In the process of further exploration of the mechanism, it was found that Notoginsenoside R1 could promote the activation of ERK1/2-P90rsk-BAD signaling pathway, and the effect of Notoginsenoside R1 was attenuated after the use of ERK1/2 inhibitor U0126. Our study indicated that the protective effects of Notoginsenoside R1 against HAPE were mainly related to the inhibition of inflammation, oxidative stress, and apoptosis. Notoginsenoside R1 may be a potential candidate for preventing HAPE.
Collapse
Affiliation(s)
- Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Yongcan Wu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing 400016, China; College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Shuiqin Li
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue, Chengdu, Sichuan 611137, China.
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, Sichuan 610075, China.
| |
Collapse
|
5
|
Shen Z, Huang D, Jia N, Zhao S, Pei C, Wang Y, Wu Y, Wang X, Shi S, Wang F, He Y, Wang Z. Protective effects of Eleutheroside E against high-altitude pulmonary edema by inhibiting NLRP3 inflammasome-mediated pyroptosis. Biomed Pharmacother 2023; 167:115607. [PMID: 37776644 DOI: 10.1016/j.biopha.2023.115607] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
Eleutheroside E (EE) is a primary active component of Acanthopanax senticosus, which has been reported to inhibit the expression of inflammatory genes, but the underlying mechanisms remain elusive. High-altitude pulmonary edema (HAPE) is a severe complication of high-altitude exposure occurring after ascent above 2500 m. However, effective and safe preventative measures for HAPE still need to be improved. This study aimed to elucidate the preventative potential and underlying mechanism of EE in HAPE. Rat models of HAPE were established through hypobaric hypoxia. Mechanistically, hypobaric hypoxia aggravates oxidative stress and upregulates (pro)-inflammatory cytokines, activating NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis, eventually leading to HAPE. EE suppressed NLRP3 inflammasome-mediated pyroptosis by inhibiting the nuclear translocation of nuclear factor kappa-Β (NF-κB), thereby protecting the lung from HAPE. However, nigericin (Nig), an NLRP3 activator, partially abolished the protective effects of EE. These findings suggest EE is a promising agent for preventing HAPE induced by NLRP3 inflammasome-mediated pyroptosis.
Collapse
Affiliation(s)
- Zherui Shen
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Demei Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Nan Jia
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Sijing Zhao
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Caixia Pei
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yilan Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yongcan Wu
- Chongqing Medical University, Chongqing 400016, China
| | - Xiaomin Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Shihua Shi
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Fei Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Yacong He
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; State Key Laboratory of Southwestern Chinese Medicine Resources School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Zhenxing Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| |
Collapse
|
6
|
Li M, Wang C, Xu WT, Zhong X. Sodium houttuyfonate plays a protective role in the asthmatic airway by alleviating the NLRP3-related pyroptosis and Th1/Th2 immune imbalance. Mol Immunol 2023; 160:103-111. [PMID: 37413910 DOI: 10.1016/j.molimm.2023.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Houttuynia cordata is an herbal compound that grows in China and exhibits anti-inflammatory, antiviral, and antioxidant properties. Additionally, pyroptosis is mediated by the activated NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome after stimulation by various inflammatory factors in asthma. OBJECTIVE To investigate the effect of sodium houttuyfonate on NLRP3 inflammasome-related pyroptosis and Th1/Th2 immune imbalance in asthma. METHODS Asthmatic mice model were made, sodium houttuyfonate was injected intraperitoneally to treat the asthmatic mice. Airway reactivity, cell classification and counting in the bronchoalveolar lavage fluid were measured. Hematoxylin-eosin and periodic acid-Schiff staining were used to analyze airway inflammation and mucus hypersecretion. Beas-2b cells were cultured, LPS, NLRP3 antagonist (Mcc950) and sodium houttuyfonate were used to intervene the Beas-2b cells, NLRP3, ASC, caspase-1, GSDMD, IL-1β, and IL-18 expression in the lung tissue and cells were analyzed using immunohistochemistry and western blot, while qRT- PCR was performed to analyze the mRNA contents in the pulmonary and the cells, respectively. Th1 and Th2 cytokines (IL-4 and IFN-γ) were detected with ELISA and the proportions of Th1 and Th2 in splenocyte were detected by flow cytometry. RESULTS Airway reactivity decreased in sodium houttuyfonate group when compared with asthmatic group mice. In the BALF, the numbers of leukocytes, eosinophils, neutrophils, lymphocytes, and macrophages were significantly lower in sodium houttuyfonate group mice than in asthmatic group mice. The proportion of TH1/TH2 cells in spleen cells and IFN-γ /IL-4 in plasma increased in sodium houttuyfonate treatment group when compared with asthma group. Immunohistochemistry, western blot and RT-PCR showed that the expressions of NLRP3, ASC, caspase-1, GSDMD, IL-1β and IL-18 were decreased in the lung tissue of mice after treated with sodium houttuyfonate when compared with those in the asthma group. However, sodium houttuyfonate combined with dexamethasone induced a stronger effect on NLRP3-related pyroptosis and Th1/Th2 immune imbalance compared to sodium houttuyfonate or dexamethasone alone. Beas-2b cells were cultured in vitro, sodium houttuyfonate can alleviate LPS-induced ASC, casepase-1, GSDMD, IL-18 and IL-1β increasing, especially in SH (10 μg/ml) treated group, but the effect less than Mcc950. CONCLUSIONS Sodium houttuyfonate can alleviated NLRP3-related pyroptosis and Th1/Th2 immune imbalance to reduce asthma airway inflammation and airway reactivity.
Collapse
Affiliation(s)
- Miao Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Chao Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Wen-Ting Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiao Zhong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
7
|
Ibrahim MA, Mohamed SR, Dkhil MA, Thagfan FA, Abdel-Gaber R, Soliman D. The effect of Moringa oleifera leaf extracts against urethane-induced lung cancer in rat model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:37280-37294. [PMID: 36567388 DOI: 10.1007/s11356-022-24813-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/13/2022] [Indexed: 06/09/2023]
Abstract
Lung cancer is one of the most common malignancies in the world, and chemotherapy can have unfavorable side effects. The aim of the present study is to evaluate the therapeutic anticancer role of Moringa oleifera leaf extracts (MLE) in urethane-induced lung cancer in adult male albino rats as compared to standard chemotherapy. Rats were categorized into four groups (10 rats/group), including negative control rats, urethane lung cancer model rats, MLE-treated lung cancer rats, and cisplatin-treated rats. Estimation of lung index, some biochemical markers of oxidative stress, quantitative real-time polymerase chain reaction (qRT-PCR), and histopathology and transmission electron microscopy were performed. The lung index was significantly increased about one-fold in urethane lung cancer model rats, but it decreased after MLE treatment. Also, MLE was able to improve the induced changes in glutathione, superoxide dismutase, and malondialdehyde concentration to be 3.8 ± 0.4 mg/g, 900.6 ± 58 U/g, and 172 ± 24 nmol/g, respectively. Additionally, after MLE treatment, the expression of EGFR-mRNA increased by about 50%. Our light and electron microscopic examination revealed that urethane group showed abnormally distributed excessive collagen fibers and the development of papillary adenocarcinoma from hyperplastic Clara cells in the lumen of terminal bronchiole with bronchiolar wall thickening, alveolar collapse, and inflammation. MLE group has moderate amount of collagen fiber and absence of tumor mass and provided more or less restoration of normal lung histology. Moreover, MLE was able to ameliorate the induced changes in mucin and PCNA positive cells in the lung by 10.8 ± 2.3%. Collectively, the current study showed that MLE could be used as anticancer agents alleviating changes associated with lung cancer in a urethane-induced lung cancer bearing rats thereby representing alternative options to toxic chemotherapy.
Collapse
Affiliation(s)
- Mona A Ibrahim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt.
| | - Sherif R Mohamed
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mohamed A Dkhil
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Felwa A Thagfan
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Rewaida Abdel-Gaber
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Doaa Soliman
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
8
|
Blockade of NLRP3/Caspase-1/IL-1β Regulated Th17/Treg Immune Imbalance and Attenuated the Neutrophilic Airway Inflammation in an Ovalbumin-Induced Murine Model of Asthma. J Immunol Res 2022; 2022:9444227. [PMID: 35664352 PMCID: PMC9159827 DOI: 10.1155/2022/9444227] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/28/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Asthma is a heterogeneous inflammatory disorder of the airways, and multiple studies have addressed the vital role of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3)/caspase-1/interleukin-1β (IL-1β) pathway in asthma, but its impact on ovalbumin- (OVA-) induced neutrophilic asthma remains unclear. Here, we explored this pathway's effect on airway inflammation in neutrophilic asthma to clarify whether blocking this signaling could alleviate asthmatic airway inflammation. Using an established OVA-induced neutrophilic asthma mouse model, we provided asthmatic mice with a highly selective NLRP3 inhibitor, MCC950, and a specific caspase-1 inhibitor, Ac-YVAD-cmk. Our results indicated that asthmatic mice exhibited increased airway hyperresponsiveness, neutrophil infiltration, and airway mucus hypersecretion, upregulated retinoid-related orphan receptor-γt (RORγt) mRNA expression, and downregulated fork head box p3 (Foxp3) mRNA expression, which was concurrent with NLRP3 inflammasome activation and upregulation of caspase-1, IL-1β, and IL-18 expression in lung. Treatment of NLRP3 inflammasome inhibitors significantly attenuated airway hyperresponsiveness, airway inflammation, and reversed T helper 17 (Th17)/regulatory T (Treg) cell imbalance in asthmatic mice. We propose that the NLRP3/caspase-1/IL-1β pathway plays an important role in the pathological process of neutrophilic asthma and provides evidence that blocking this pathway could potentially be a treatment strategy to ameliorate airway inflammation in asthma after validation with future experimental and clinical studies.
Collapse
|
9
|
Zafar MS, Shahid K, Gobe GC, Yasmin R, Naseem N, Shahzad M. Suppression of cytokine storm and associated inflammatory mediators by salicylaldehyde derivative of pregabalin: An innovative perspective for alleviating airway inflammation and lung remodeling. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2022; 34:101877. [DOI: 10.1016/j.jksus.2022.101877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
|
10
|
Amelioration of Ovalbumin-Induced Allergic Asthma by Juglans regia via Downregulation of Inflammatory Cytokines and Upregulation of Aquaporin-1 and Aquaporin-5 in Mice. J Trop Med 2022; 2022:6530095. [PMID: 35401757 PMCID: PMC8986429 DOI: 10.1155/2022/6530095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/22/2022] [Accepted: 03/10/2022] [Indexed: 11/17/2022] Open
Abstract
Juglans regia (J. regia) has been used traditionally to treat cough and asthma. The present study evaluates the immunomodulatory and anti-inflammatory potential of J. regia against ovalbumin (OVA)-induced allergic asthma. Intraperitoneal sensitization proceeded by intranasal challenge with OVA was used to induce allergic asthma. BALB/c mice were treated with methanol, n-hexane, and ethyl acetate extracts of J. regia and methylprednisolone one week after 2nd sensitization with OVA and continued for 7 days. mRNA expression levels of IL-4, IL-5, IL-13, AQP-1, AQP-5 TNF-α, TGF-β, and NF-kB were determined using reverse transcription polymerase chain reaction. Hematoxylin and eosin, and periodic acidic-Schiff stains were used for histopathological studies of lung tissues. The data presented all three extracts of J. regia significantly ameliorated airway inflammation by reducing expression levels of IL-4, IL-5, and IL-13 and TNF-α in OVA-treated mice. The suppression of goblet cells hyperplasia and inflammatory cells infiltration by J. regia involved low TGF-β and NF-kB levels. Pretreatment with J. regia also increased the AQP-1 and AQP-5 expression levels in mice treated with OVA. This study supported the traditional use of J. regia and proposed that J. regia ameliorated allergic asthma by suppression of proinflammatory cytokines and elevation of AQP-1 and AQP-5 expression levels.
Collapse
|
11
|
Polystichum braunii ameliorates airway inflammation by attenuation of inflammatory and oxidative stress biomarkers, and pulmonary edema by elevation of aquaporins in ovalbumin-induced allergic asthmatic mice. Inflammopharmacology 2022; 30:639-653. [PMID: 35257281 DOI: 10.1007/s10787-022-00944-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/09/2022] [Indexed: 11/05/2022]
Abstract
Asthma is a chronic inflammation of pulmonary airways associated with bronchial hyper-responsiveness. The study was aimed to validate the folkloric use of Polystichum braunii (PB) against ovalbumin (OVA)-induced asthmatic and chemical characterization OF both extracts. Allergic asthma was developed by intraperitoneal sensitization with an OVA on days 1 and 14 followed by intranasal challenge. Mice were treated with PB methanolic (PBME) and aqueous extract (PBAE) orally at 600, 300, and 150 mg/kg and using dexamethasone (2 mg/kg) as standard from day 15 to 26. High performance liquid chromatography-diode array detector analysis revealed the presence of various bioactive compounds such as catechin, vanillic acid, and quercetin. The PBME and PBAE profoundly (p < 0.0001-0.05) declined immunoglobulin E level, lungs wet/dry weight ratio, and total and differential leukocyte count in blood and bronchial alveolar lavage fluid of treated mice in contrast to disease control. Histopathological examination showed profoundly decreased inflammatory cell infiltration and goblet cell hyperplasia in treated groups. Both extracts caused significant (p < 0.0001-0.05) diminution of IL-4, IL-5, IL-13, IL-6, IL-1β, TNF-α, and NF-κB and upregulation of aquaporins (1 and 5), which have led to the amelioration of pulmonary inflammation and attenuation of lung edema in treated mice. Both extracts profoundly (p < 0.0001-0.05) restored the activities of SOD, CAT, GSH and reduced the level of MDA dose dependently. Both extracts possessed significant anti-asthmatic action mainly PBME 600 mg/kg might be due to phenols and flavonoids and could be used as a potential therapeutic option in the management of allergic asthma.
Collapse
|
12
|
Wagner K, Unger L, Salman MM, Kitchen P, Bill RM, Yool AJ. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int J Mol Sci 2022; 23:1388. [PMID: 35163313 PMCID: PMC8836214 DOI: 10.3390/ijms23031388] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The aquaporins (AQPs) are a family of small integral membrane proteins that facilitate the bidirectional transport of water across biological membranes in response to osmotic pressure gradients as well as enable the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body. Here, we review their key roles in fluid homeostasis, glandular secretions, signal transduction and sensation, barrier function, immunity and inflammation, cell migration, and angiogenesis. Evidence from a wide variety of studies now supports a view of the functions of AQPs being much more complex than simply mediating the passive flow of water across biological membranes. The discovery and development of small-molecule AQP inhibitors for research use and therapeutic development will lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.
Collapse
Affiliation(s)
- Kim Wagner
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Mootaz M. Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Roslyn M. Bill
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
13
|
Hu Y, Sun J, Wang T, Wang H, Zhao C, Wang W, Yan K, Yan X, Sun H. Compound Danshen Dripping Pill inhibits high altitude-induced hypoxic damage by suppressing oxidative stress and inflammatory responses. PHARMACEUTICAL BIOLOGY 2021; 59:1585-1593. [PMID: 34808069 PMCID: PMC8635678 DOI: 10.1080/13880209.2021.1998139] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Previous studies indicate that compound Danshen Dripping Pill (CDDP) improves the adaptation to high-altitude exposure. However, its mechanism of action is not clear. OBJECTIVE To explore the protective effect of CDDP on hypobaric hypoxia (HH) and its possible mechanism. MATERIALS AND METHODS A meta-analysis of 1051 human volunteers was performed to evaluate the effectiveness of CDDP at high altitudes. Male Sprague-Dawley rats were randomized into 5 groups (n = 6): control at normal pressure, model, CDDP-170 mg/kg, CDDP-340 mg/kg and acetazolamide groups. HH was simulated at an altitude of 5500 m for 24 h. Animal blood was collected for arterial blood-gas analysis and cytokines detection and their organs were harvested for pathological examination. Expression levels of AQP1, NF-κB and Nrf2 were determined by immunohistochemical staining. RESULTS The meta-analysis data indicated that the ratio between the combined RR of the total effective rate and the 95% CI was 0.23 (0.06, 0.91), the SMD and 95% CI of SO2 was 0.37 (0.12, 0.62). Pre-treatment of CDDP protected rats from HH-induced pulmonary edoema and heart injury, left-shifted oxygen-dissociation curve and decreased P50 (30.25 ± 3.72 vs. 37.23 ± 4.30). Mechanistically, CDDP alleviated HH-reinforced ROS by improving SOD and GPX1 while inhibiting pro-inflammatory cytokines and NF-κB expression. CDDP also decreased HH-evoked D-dimer, erythrocyte aggregation and blood hemorheology, promoting AQP1 and Nrf2 expression. DISCUSSION AND CONCLUSIONS Pre-treatment with CDDP could prevent HH-induced tissue damage, oxidative stress and inflammatory response. Suppressed NF-κB and up-regulated Nrf2 might play significant roles in the mechanism of CDDP.
Collapse
Affiliation(s)
- Yunhui Hu
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Jia Sun
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Tongxing Wang
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Hairong Wang
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Chunlai Zhao
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Wenjia Wang
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
| | - Kaijing Yan
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
- The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China
- Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - Xijun Yan
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
- The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China
- Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| | - He Sun
- GeneNet Pharmaceuticals Co. Ltd, Tianjin, P.R. China
- The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China
- Tasly Pharmaceutical Group Co., Ltd, Tianjin, China
| |
Collapse
|
14
|
Piccapane F, Gerbino A, Carmosino M, Milano S, Arduini A, Debellis L, Svelto M, Caroppo R, Procino G. Aquaporin-1 Facilitates Transmesothelial Water Permeability: In Vitro and Ex Vivo Evidence and Possible Implications in Peritoneal Dialysis. Int J Mol Sci 2021; 22:12535. [PMID: 34830416 PMCID: PMC8622642 DOI: 10.3390/ijms222212535] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
We previously showed that mesothelial cells in human peritoneum express the water channel aquaporin 1 (AQP1) at the plasma membrane, suggesting that, although in a non-physiological context, it may facilitate osmotic water exchange during peritoneal dialysis (PD). According to the three-pore model that predicts the transport of water during PD, the endothelium of peritoneal capillaries is the major limiting barrier to water transport across peritoneum, assuming the functional role of the mesothelium, as a semipermeable barrier, to be negligible. We hypothesized that an intact mesothelial layer is poorly permeable to water unless AQP1 is expressed at the plasma membrane. To demonstrate that, we characterized an immortalized cell line of human mesothelium (HMC) and measured the osmotically-driven transmesothelial water flux in the absence or in the presence of AQP1. The presence of tight junctions between HMC was investigated by immunofluorescence. Bioelectrical parameters of HMC monolayers were studied by Ussing Chambers and transepithelial water transport was investigated by an electrophysiological approach based on measurements of TEA+ dilution in the apical bathing solution, through TEA+-sensitive microelectrodes. HMCs express Zo-1 and occludin at the tight junctions and a transepithelial vectorial Na+ transport. Real-time transmesothelial water flux, in response to an increase of osmolarity in the apical solution, indicated that, in the presence of AQP1, the rate of TEA+ dilution was up to four-fold higher than in its absence. Of note, we confirmed our data in isolated mouse mesentery patches, where we measured an AQP1-dependent transmesothelial osmotic water transport. These results suggest that the mesothelium may represent an additional selective barrier regulating water transport in PD through functional expression of the water channel AQP1.
Collapse
Affiliation(s)
- Francesca Piccapane
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy;
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Arduino Arduini
- Department of Research and Development, CoreQuest Sagl, 6900 Lugano, Switzerland;
| | - Lucantonio Debellis
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Rosa Caroppo
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (F.P.); (A.G.); (S.M.); (L.D.); (M.S.); (R.C.)
| |
Collapse
|
15
|
Chen L, Xu W, Mao S, Zhu R, Zhang J. Autoantibody of interleukin-17A induced by recombinant Mycobacterium smegmatis attenuates airway inflammation in mice with neutrophilic asthma. J Asthma 2021; 59:2117-2126. [PMID: 34644222 DOI: 10.1080/02770903.2021.1989696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Previous studies have shown Interleukin (IL)-17A as an important contributor to the development of severe asthma, which is mainly characterized by neutrophilic inflammation and less response to corticosteroids. Consequently, the IL-17A-neutrophil axis could be a potential therapeutic target. Previously, we constructed a recombinant Mycobacterium smegmatis (rMS) expressing fusion protein Ag85A-IL-17A, and confirmed it could induce production of IL-17A autoantibody in vivo. This study uses a murine model of neutrophilic asthma to further investigate the effects of rMS on airway inflammation. METHODS DO11.10 mice were divided into four groups: phosphate buffered saline (PBS), asthma, rMS and MS. This murine model of neutrophilic asthma was established with ovalbumin (OVA) challenge, whereby PBS, rMS and MS were administered intranasally. Anti-inflammatory effects on inflammatory cell infiltration and expression of inflammatory mediators in bronchoalveolar lavage fluid (BALF) were evaluated, along with histopathological changes in lung tissues. RESULTS A sustained high-titer IL-17A autoantibody was detected in sera of the rMS group. Compared to the asthma group, the number of neutrophils, IL-17A, CXCL-1 levels and MPO activity in the rMS group were all significantly reduced (p < 0.01). Histological analysis showed rMS remarkably suppressed inflammatory infiltration around bronchia. The inflammation score and the mucus score in the rMS group were both significantly lower than those in the asthma group (p < 0.001). CONCLUSION rMS ameliorated airway inflammation in mice with neutrophilic asthma caused by inducing IL-17A autoantibody and regulating the IL-17A-neutrophil axis, thus offering a possible novel treatment for neutrophilic asthma.
Collapse
Affiliation(s)
- Ling Chen
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wanting Xu
- Department of Neonatology, Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Song Mao
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ruochen Zhu
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jianhua Zhang
- Department of Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Tardelli M, Stulnig TM. Aquaporin regulation in metabolic organs. VITAMINS AND HORMONES 2021; 112:71-93. [PMID: 32061350 DOI: 10.1016/bs.vh.2019.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aquaporins (AQPs) are a family of 13 small trans-membrane proteins, which facilitate shuttling of glycerol, water and urea. The peculiar role of AQPs in glycerol transport makes them attractive targets in metabolic organs since glycerol represents the backbone of triglyceride synthesis. Importantly, AQPs are known to be regulated by various nuclear receptors which in turn govern lipid and glucose metabolism as well as inflammatory cascades. Here, we review the role of AQPs regulation in metabolic organs exploring their physiological impact in health and disease.
Collapse
Affiliation(s)
- Matteo Tardelli
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Cornell Department of Medicine, Weill Cornell Medical College, New York, NY, United States; Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas M Stulnig
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
17
|
Molecular mechanisms of An-Chuan Granule for the treatment of asthma based on a network pharmacology approach and experimental validation. Biosci Rep 2021; 41:228000. [PMID: 33645621 PMCID: PMC7990088 DOI: 10.1042/bsr20204247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
An-Chuan Granule (ACG), a traditional Chinese medicine (TCM) formula, is an effective treatment for asthma but its pharmacological mechanism remains poorly understood. In the present study, network pharmacology was applied to explore the potential mechanism of ACG in the treatment of asthma. The tumor necrosis factor (TNF), Toll-like receptor (TLR), and Th17 cell differentiation-related, nucleotide-binding oligomerization domain (NOD)-like receptor, and NF-kappaB pathways were identified as the most significant signaling pathways involved in the therapeutic effect of ACG on asthma. A mouse asthma model was established using ovalbumin (OVA) to verify the effect of ACG and the underlying mechanism. The results showed that ACG treatment not only attenuated the clinical symptoms, but also reduced inflammatory cell infiltration, mucus secretion and MUC5AC production in lung tissue of asthmatic mice. In addition, ACG treatment notably decreased the inflammatory cell numbers in bronchoalveolar lavage fluid (BALF) and the levels of pro-inflammatory cytokines (including IL-6, IL-17, IL-23, TNF-alpha, IL-1beta and TGF-beta) in lung tissue of asthmatic mice. In addition, ACG treatment remarkably down-regulated the expression of TLR4, p-P65, NLRP3, Caspase-1 and adenosquamous carcinoma (ASC) in lung tissue. Further, ACG treatment decreased the expression of receptor-related orphan receptor (RORγt) in lung tissue but increased that of Forkhead box (Foxp3). In conclusion, the above results demonstrate that ACG alleviates the severity of asthma in a ´multi-compound and multi-target’ manner, which provides a basis for better understanding of the application of ACG in the treatment of asthma.
Collapse
|
18
|
Amelioration of airway inflammation and pulmonary edema by Teucrium stocksianum via attenuation of pro-inflammatory cytokines and up-regulation of AQP1 and AQP5. Respir Physiol Neurobiol 2020; 284:103569. [PMID: 33144273 DOI: 10.1016/j.resp.2020.103569] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 01/21/2023]
Abstract
Current study investigates the immunomodulatory effects of T. stocksianum using mouse model of ovalbumin (OVA)-induced allergic asthma. The mice were treated with methanolic extract, n-hexane, and ethyl acetate fractions for consecutive 7 days along with intranasal challenge. The mRNA expression levels of interleukin-4 (IL-4), IL-5, Aquaporin-1 (AQP1) and Aquaporin-5 (AQP5) were evaluated using reverse transcription polymerase chain reaction. The data showed that T. stocksianum significantly reduced airway inflammation as indicated by reduced inflammatory cell infiltration in lungs, and attenuated total and differential leukocyte counts both in blood and BALF. Expression levels of pro-inflammatory IL-4 and IL-5 in lungs were also found significantly reduced. T. stocksianum significantly reduced pulmonary edema as indicated by reduced lung wet/dry ratio and goblet cell hyperplasia. AQP1 and AQP5 expression levels were also found elevated in treatment groups. In conclusion, T. stocksianum possesses anti-asthmatic activity which may be attributed to reduction in IL-4 and IL-5 expression levels, and elevation in AQP1 and AQP5 expression levels.
Collapse
|
19
|
Yadav E, Yadav N, Hus A, Yadav JS. Aquaporins in lung health and disease: Emerging roles, regulation, and clinical implications. Respir Med 2020; 174:106193. [PMID: 33096317 DOI: 10.1016/j.rmed.2020.106193] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/17/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Aquaporins (AQPs) aka water channels are a family of conserved transmembrane proteins (~30 kDa monomers) expressed in various organ systems. Of the 13 AQPs (AQP0 through AQP12) in the human body, four (AQPs 1, 3, 4, and 5) are expressed in the respiratory system. These channels are conventionally known for mediating transcellular fluid movements. Certain AQPs (aquaglyceroporins) have the capability to transport glycerol and potentially other solutes. There is an emerging body of literature unveiling the non-conventional roles of AQPs such as in cell proliferation and migration, gas permeation, signal potentiation, etc. Initial gene knock-out studies established a physiological role for lung AQPs, particularly AQP5, in maintaining homeostasis, by mediating fluid secretion from submucosal glands onto the airway surface liquid (ASL) lining. Subsequent studies have highlighted the functional significance of AQPs, particularly AQP1 and AQP5 in lung pathophysiology and diseases, including but not limited to chronic and acute lung injury, chronic obstructive pulmonary disease (COPD), other inflammatory lung conditions, and lung cancer. AQP1 has been suggested as a potential prognostic marker for malignant mesothelioma. Recent efforts are directed toward exploiting AQPs as targets for diagnosis, prevention, intervention, and/or treatment of various lung conditions. Emerging information on regulatory pathways and directed mechanistic research are posited to unravel novel strategies for these clinical implications. Future considerations should focus on development of AQP inhibitors, blockers, and modulators for therapeutic needs, and better understanding the role of lung-specific AQPs in inter-individual susceptibility to chronic lung diseases such as COPD and cancer.
Collapse
Affiliation(s)
- Ekta Yadav
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Niket Yadav
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA, 22908-0738, USA
| | - Ariel Hus
- Department of Biology, University of Miami, Coral Gables, Florida, 33146, USA
| | - Jagjit S Yadav
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
20
|
Mariajoseph-Antony LF, Kannan A, Panneerselvam A, Loganathan C, Anbarasu K, Prahalathan C. Could aquaporin modulators be employed as prospective drugs for COVID-19 related pulmonary comorbidity? Med Hypotheses 2020; 143:110201. [PMID: 33017909 PMCID: PMC7430244 DOI: 10.1016/j.mehy.2020.110201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/15/2020] [Indexed: 01/03/2023]
Abstract
COVID-19 initially an epidemic caused by SARS-CoV-2 has turned out to be a life- threatening global pandemic with increased morbidity and mortality. The presence of cytokine storm has been linked with the pathogenesis of severe lung injury as evinced in COVID-19. Aquaporins (AQPs) are molecular water channels, facilitating water transport across the cell membrane in response to osmotic gradients. Impairment in alveolar fluid clearance due to altered functional expression of respiratory AQPs highlight their pathophysiological significance in pulmonary edema associated respiratory illness. Therefore, we hypothesize that targeted modulation of AQPs in lungs in the intervening period of time, could diminish the dreadful effects of inflammation- induced comorbidity in COVID-19.
Collapse
Affiliation(s)
- Lezy Flora Mariajoseph-Antony
- Molecular Endocrinology Laboratory, Department of Biochemistry, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Arun Kannan
- Molecular Endocrinology Laboratory, Department of Biochemistry, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Antojenifer Panneerselvam
- Molecular Endocrinology Laboratory, Department of Biochemistry, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Chithra Loganathan
- Molecular Endocrinology Laboratory, Department of Biochemistry, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Kumarasamy Anbarasu
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Chidambaram Prahalathan
- Molecular Endocrinology Laboratory, Department of Biochemistry, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, India.
| |
Collapse
|
21
|
Sevoflurane modulates AQPs (1,5) expression and endoplasmic reticulum stress in mice lung with allergic airway inflammation. Biosci Rep 2020; 39:221068. [PMID: 31710085 PMCID: PMC6879378 DOI: 10.1042/bsr20193282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/29/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023] Open
Abstract
Sevoflurane was found to show protective roles in mice with asthma, however, the mechanism of which needs further exploring. Aquaporins (AQPs) have been demonstrated to be involved in the pathogenesis of asthma, while endoplasmic reticulum stress has been reported to be related to many inflammatory diseases and involved in protein processing, including AQPs. The present study aimed to determine the role of sevoflurane in AQPs (AQP1,3,4,5) expression in mice with allergic airway inflammation and the probable mechanism. The increased number of inflammatory cells infiltrating the lung tissue, and the elevated levels of tumor necrosis factor-α (TNF-α) and interleukin (IL) 13 (IL-13) were all decreased after sevoflurane treatment (all P<0.05). Meanwhile, mRNA levels of AQP1 and AQP5 but not AQP3 and AQP4 were decreased in ovalbumin (OVA)-induced allergic mice lung. Both the decreased mRNA expression and protein levels of AQP1 and AQP5 in allergic lung tissues were reversed by sevoflurane treatment. Furthermore, we established that sevoflurane inhibited the OVA-induced protein increase in the endoplasmic reticulum (ER) stress markers BiP and C/EBP homologous protein (CHOP). Collectively, these findings suggested that sevoflurane modulated the expression and protein level of AOPs (AQP1, AQP5) as well as inhibited ER stress response in OVA-induced allergic airway inflammation of mice.
Collapse
|
22
|
Abstract
Aquaporins (AQPs) are transmembrane channel proteins that mainly facilitate the water translocation through the plasma cell membrane. For several years these proteins have been extensively examined for their biologic role in health and their potential implication in different diseases. Technological improvements associated with the methods employed to evaluate the functions of the AQPs have provided us with significant new knowledge. In this chapter, we will examine the role of AQPs in health and disease based on the latest currently available evidence.
Collapse
Affiliation(s)
- Dimitrios E Magouliotis
- Division of Surgery and Interventional Sciences, UCL, London, United Kingdom; Department of Surgery, University of Thessaly, Biopolis, Larissa, Greece.
| | | | - Alexis A Svokos
- Geisinger Lewisburg-Women's Health, Lewisburg, PA, United States
| | - Konstantina A Svokos
- The Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
23
|
Immunomodulatory and Anti-Inflammatory Potential of Curcumin for the Treatment of Allergic Asthma: Effects on Expression Levels of Pro-inflammatory Cytokines and Aquaporins. Inflammation 2020; 42:2037-2047. [PMID: 31407145 DOI: 10.1007/s10753-019-01066-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Curcumin is well known for possessing anti-inflammatory properties and for its beneficial effects in the treatment of asthma. Current study investigates the immunomodulatory and anti-inflammatory effects of curcumin using mouse model of ovalbumin-induced allergic asthma. BALB/c mice were immunized with ovalbumin on day 0 and 14 to induce allergic asthma. Animals were treated with two different doses of curcumin (20 mg/kg and 100 mg/kg) and methylprednisolone from day 21 to 28. Mice were also daily challenged intranasally with ovalbumin during treatment period, and all groups were sacrificed at day 28. Histopathological examination showed amelioration of allergic asthma in treated groups as evident by the attenuation of infiltration of inflammatory cells, goblet cell hyperplasia, alveolar thickening, and edema and vascular congestion. Curcumin significantly reduced total and differential leukocyte counts in both bronchoalveolar lavage fluid and blood. Reverse transcription polymerase chain reaction analysis showed significantly suppressed mRNA expression levels of IL-4 and IL-5 (pro-inflammatory cytokines), TNF-α, TGF-β (pro-fibrotic cytokines), eotaxin (chemokine), and heat shock protein 70 (marker of airway obstruction) in treated groups. Attenuation of these pro-inflammatory markers might have led to the suppression of airway inflammation. The expression levels of aquaporin-1 (AQP) and AQP-5 were found significantly elevated in experimental groups which might be responsible for reduction of pulmonary edema. In conclusion, curcumin significantly ameliorated allergic asthma. The anti-asthmatic effect might be attributed to the suppression of pro-inflammatory cytokines, and elevation of aquaporin expression levels, suggesting further studies and clinical trials to establish its candidature in the treatment of allergic asthma.
Collapse
|
24
|
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: 46] [Impact Index Per Article: 7.7] [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
|
25
|
Ahsan F, Shabbir A, Shahzad M, Mobashar A, Sharif M, Basheer MI, Tareen RB, Syed NIH. Amelioration of allergic asthma by Ziziphora clinopodioides via upregulation of aquaporins and downregulation of IL4 and IL5. Respir Physiol Neurobiol 2019; 266:39-46. [PMID: 31015030 DOI: 10.1016/j.resp.2019.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/29/2019] [Accepted: 04/10/2019] [Indexed: 01/26/2023]
Abstract
Ziziphora clinopodioides has been frequently used as an anti asthmatic plant in traditional medication. Recent work explores the anti-asthmatic activity of Z. clinopodioides in allergen-induced asthmatic mice. Intraperitoneal sensitization followed by intranasal challenge were given with ovalbumin (allergen) to develop allergic asthma. Investigational groups of animals were administered with drug methylprednisolone (MP) (15 mg/kg body weight), n-hexane fraction, ethylacetate fraction, and methanolic extract of Z. clinopodioides extract (500 mg/kg b.w.) for successive 07 days. Hematoxyline and eosin (H&E) and periodic acid-Schiff (PAS) stains were used to evaluate histopathological parameters on lung tissues. As an index of lungs tissues edema, wet/dry weight ratio of lungs was determined. Evaluation of expression levels of AQP1, AQP5, IL4, and IL5 was conducted by using RT-PCR. The data exhibited that both Z. clinopodioides and MP attenuated differential and total leukocyte counts in hematological examination i.e. in BALF and blood. Treatment with Z. clinopodioides also caused suppression of inflammatory cell infiltration and expression levels of IL4 and IL5, the later could have caused attenuation of pulmonary inflammation. The study also found decline in lung wet/dry ratio and goblet cellh hyperplasia in treated groups which indicates amelioration of lung edema. Treatment with Z. clinopodioides significantly increased the expression levels of aquaporin-1 and -5, which could have led to reduction in lung edema. The treatment with MP showed comparable results to Z. clinopodioides. Current investigation revealed that Z. clinopodioides possessed anti-asthmatic property which might be accredited to upregulagted AQP1 and AQP5 levels and downregulated IL4 and IL5 levels.
Collapse
Affiliation(s)
- Fatima Ahsan
- Pharmacology section, Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Arham Shabbir
- Pharmacology section, Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan; Department of Pharmacy, The University of Lahore-Gujrat Campus, Gujrat, Pakistan.
| | - Muhammad Shahzad
- Department of Pharmacology, The University of Health Sciences, Lahore, Pakistan
| | - Aisha Mobashar
- Pharmacology section, Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Marriam Sharif
- Pharmacology section, Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | | | | | | |
Collapse
|
26
|
Ikarashi N, Nagoya C, Kon R, Kitaoka S, Kajiwara S, Saito M, Kawabata A, Ochiai W, Sugiyama K. Changes in the Expression of Aquaporin-3 in the Gastrointestinal Tract Affect Drug Absorption. Int J Mol Sci 2019; 20:ijms20071559. [PMID: 30925715 PMCID: PMC6479729 DOI: 10.3390/ijms20071559] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Aquaporin-3 (AQP3) plays an important role in water transport in the gastrointestinal (GI) tract. In this study, we conducted a Caco-2 cell permeability assay to examine how changes in the expression and function of AQP3 affect the rate at which a drug is absorbed via passive transport in the GI tract. When the function of AQP3 was inhibited by mercuric chloride or phloretin, there was no change in warfarin permeability. In contrast, when the expression of AQP3 protein was decreased by prostaglandin E₂ (PGE₂) treatment, warfarin permeability increased to approximately twice the control level, and membrane fluidity increased by 15%. In addition, warfarin permeability increased to an extent comparable to that after PGE₂ treatment when cell membrane fluidity was increased by 10% via boric acid/EDTA treatment. These findings suggest the possibility that the increased drug absorption under decreased AQP3 expression was attributable to increased membrane fluidity. The results of this study demonstrate that the rate of water transport has little effect on drug absorption. However, our findings also indicate that although AQP3 and other similar transmembrane proteins do not themselves transport drugs, changes in their expression levels can cause changes in cell membrane fluidity, thus affecting drug absorption rates.
Collapse
Affiliation(s)
- Nobutomo Ikarashi
- Department of Biomolecular Pharmacology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Chika Nagoya
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Risako Kon
- Department of Biomolecular Pharmacology, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Satoshi Kitaoka
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Sayuri Kajiwara
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Masayo Saito
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Akane Kawabata
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Wataru Ochiai
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | - Kiyoshi Sugiyama
- Department of Functional Molecular Kinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| |
Collapse
|
27
|
Wittekindt OH, Dietl P. Aquaporins in the lung. Pflugers Arch 2018; 471:519-532. [PMID: 30397774 PMCID: PMC6435619 DOI: 10.1007/s00424-018-2232-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/04/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022]
Abstract
The lung is the interface between air and blood where the exchange of oxygen and carbon dioxide occurs. The surface liquid that is directly exposed to the gaseous compartment covers both conducting airways and respiratory zone and forms the air-liquid interface. The barrier that separates this lining fluid of the airways and alveoli from the extracellular compartment is the pulmonary epithelium. The volume of the lining fluid must be kept in a range that guarantees an appropriate gas exchange and other functions, such as mucociliary clearance. It is generally accepted that this is maintained by balancing resorptive and secretory fluid transport across the pulmonary epithelium. Whereas osmosis is considered as the exclusive principle of fluid transport in the airways, filtration may contribute to alveolar fluid accumulation under pathologic conditions. Aquaporins (AQP) facilitate water flux across cell membranes, and as such, they provide a transcellular route for water transport across epithelia. However, their contribution to near-isosmolar fluid conditions in the lung still remains elusive. Herein, we discuss the role of AQPs in the lung with regard to fluid homeostasis across the respiratory epithelium.
Collapse
Affiliation(s)
- Oliver H Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| | - Paul Dietl
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| |
Collapse
|
28
|
Tardelli M, Claudel T, Bruschi FV, Trauner M. Nuclear Receptor Regulation of Aquaglyceroporins in Metabolic Organs. Int J Mol Sci 2018; 19:E1777. [PMID: 29914059 PMCID: PMC6032257 DOI: 10.3390/ijms19061777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Nuclear receptors, such as the farnesoid X receptor (FXR) and the peroxisome proliferator-activated receptors gamma and alpha (PPAR-γ, -α), are major metabolic regulators in adipose tissue and the liver, where they govern lipid, glucose, and bile acid homeostasis, as well as inflammatory cascades. Glycerol and free fatty acids are the end products of lipid droplet catabolism driven by PPARs. Aquaporins (AQPs), a family of 13 small transmembrane proteins, facilitate the shuttling of water, urea, and/or glycerol. The peculiar role of AQPs in glycerol transport makes them pivotal targets in lipid metabolism, especially considering their tissue-specific regulation by the nuclear receptors PPARγ and PPARα. Here, we review the role of nuclear receptors in the regulation of glycerol shuttling in liver and adipose tissue through the function and expression of AQPs.
Collapse
Affiliation(s)
- Matteo Tardelli
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Francesca Virginia Bruschi
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| |
Collapse
|
29
|
Liu X, Shen J, Fan D, Qiu X, Guo Q, Zheng K, Luo H, Shu J, Lu C, Zhang G, Lu A, Ma C, He X. Yupingfeng San Inhibits NLRP3 Inflammasome to Attenuate the Inflammatory Response in Asthma Mice. Front Pharmacol 2017; 8:944. [PMID: 29311942 PMCID: PMC5743824 DOI: 10.3389/fphar.2017.00944] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 11/13/2022] Open
Abstract
Yupingfeng San (YPFS) is a representative Traditional Chinese Medicine (TCM) formula with accepted therapeutic effect on Asthma. However, its action mechanism is still obscure. In this study, we used network pharmacology to explore potential mechanism of YPFS on asthma. Nucleotide-binding oligomerization domain (NOD)-like receptor pathway was shown to be the top one shared signaling pathway associated with both YPFS and asthma. In addition, NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome was treated as target protein in the process of YPFS regulating asthma. Further, experimental validation was done by using LPS-stimulated U937 cells and ovalbumin (OVA)-sensitized BALB/c mice model. In vitro experiments showed that YPFS significantly decreased the production of TNF-α and IL-6, as well as both mRNA and protein levels of IL-1β, NLRP3, Caspase-1 and ASC in LPS-stimulated U937 cells. In vivo experiment indicated that YPFS treatment not only attenuated the clinical symptoms, but also reduced inflammatory cell infiltration, mucus secretion and MUC5AC production in lung tissue of asthmatic mice. Moreover, YPFS treatment remarkably decreased the mRNA and protein levels of IL-1β, NLRP3, Caspase-1 and ASC in lung tissue of asthmatic mice. In conclusion, these results demonstrated that YPFS could inhibit NLRP3 inflammasome components to attenuate the inflammatory response in asthma.
Collapse
Affiliation(s)
- Xue Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jiawen Shen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Danping Fan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuemei Qiu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Qingqing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Kang Zheng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hui Luo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jun Shu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.,School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chaoying Ma
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.,Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| |
Collapse
|
30
|
Chang YL, Jian KR, Lin CS, Wang HW, Liu SC. Dexamethasone attenuates methacholine-mediated aquaporin 5 downregulation in human nasal epithelial cells via suppression of NF-κB activation. Int Forum Allergy Rhinol 2017; 8:64-71. [PMID: 29083535 DOI: 10.1002/alr.22035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/05/2017] [Accepted: 10/08/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cholinergic stimulation plays a major role in inflammatory airway diseases. However, its role in airway surface liquid homeostasis and aquaporin 5 (AQP5) regulation remains unclear. In this study we sought to determine the effects of methacholine and dexamethasone on AQP5 expression in human nasal epithelial cells (HNEpC). METHODS HNEpC were cultured with methacholine or dexamethasone at 4 concentrations in vitro. The subcellular distribution of AQP5 was explored using immunocytochemistry. The pharmacologic effects of methacholine and dexamethasone on the expression of the phosphorylation of cyclic adenosine monophosphate-responsive element binding protein (p-CREB), AQP5, and nuclear factor-kappaB (NF-κB) were examined using Western blotting. RESULTS AQP5 was found to be located in cell membrane and cytoplasm and present in every group without a statistically significant difference. Methacholine inhibited expression of AQP5 and p-CREB in HNEpC, whereas dexamethasone increased these protein levels dose-dependently in a statistically significant manner. In turn, HNEpC treated with methacholine and dexamethasone showed the same trends as those intervened separately with these 2 drugs. Moreover, dexamethasone had the ability to reverse the inhibitory effect of methacholine. Western blotting revealed that, after incubation with 10-4 mol/L methacholine, NF-κB increased significantly, by 186.67%, compared with the untreated control group. Again, such an increase could be significantly reversed after dexamethasone treatment. CONCLUSION NF-κB activation is important for inhibition of p-CREB/AQP5 expression after methacholine intervention, and dexamethasone adjusts it to the opposite side. This observation could provide additional insight into the anti-inflammatory effects of glucocorticoids that contribute to maintaining airway surface liquid and mucosal defense.
Collapse
Affiliation(s)
- Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Kai Ren Jian
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Hsing-Won Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center.,Department of Otolaryngology-Head and Neck Surgery, Shuang Ho Hospital, Taipei, Taiwan, Republic of China
| | - Shao-Cheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center
| |
Collapse
|
31
|
Abstract
Fibromyalgia appears to present in subgroups with regard to biological pain induction, with primarily inflammatory, neuropathic/neurodegenerative, sympathetic, oxidative, nitrosative, or muscular factors and/or central sensitization. Recent research has also discussed glial activation or interrupted dopaminergic neurotransmission, as well as increased skin mast cells and mitochondrial dysfunction. Therapy is difficult, and the treatment options used so far mostly just have the potential to address only one of these aspects. As ambroxol addresses all of them in a single substance and furthermore also reduces visceral hypersensitivity, in fibromyalgia existing as irritable bowel syndrome or chronic bladder pain, it should be systematically investigated for this purpose. Encouraged by first clinical observations of two working groups using topical or oral ambroxol for fibromyalgia treatments, the present paper outlines the scientific argument for this approach by looking at each of the aforementioned aspects of this complex disease and summarizes putative modes of action of ambroxol. Nevertheless, at this point the evidence basis for ambroxol is not strong enough for clinical recommendation.
Collapse
Affiliation(s)
- Kai-Uwe Kern
- Institute of Pain Medicine/Pain Practice, Wiesbaden, Germany
| | | |
Collapse
|
32
|
[More than expectorant: new scientific data on ambroxol in the context of the treatment of bronchopulmonary diseases]. MMW Fortschr Med 2017. [PMID: 28643291 DOI: 10.1007/s15006-017-9805-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ambroxol has been established for decades in the treatment of acute and chronic respiratory diseases. In 2015, the European Medicines Agency reassessed the clinical benefit-risk ratio of the drug. OBJECTIVE What new scientific data on ambroxol, which are relevant to the treatment of bronchopulmonary diseases, are available? METHOD The review is based on a systematic literature research in medline with the search term "ambroxol" during the publication period 2006-2015. Non-relevant publications were excluded manually. RESULTS AND CONCLUSIONS Ambroxol is still intensively researched. The traditional indication as an expectorant is confirmed. But there is also an ever better understanding of the various mechanisms of action as well as the ever more exact modeling of the structures under investigation. New fields of application are conceivable, e. g. in patients with severe pulmonary disease who undergo surgery or who are in intensive care, as an adjuvant in anti-infective therapies, especially in infections with biofilm-producing pathogens, or in rare diseases such as lysosomal storage diseases. However, final evidence of the clinical relevance in these fields of application is still missing.
Collapse
|
33
|
Kamalaldin NA, Sulaiman SA, Yusop MR, Yahaya B. Does Inhalation of Virgin Coconut Oil Accelerate Reversal of Airway Remodelling in an Allergic Model of Asthma? Int J Inflam 2017; 2017:8741851. [PMID: 28660089 PMCID: PMC5474257 DOI: 10.1155/2017/8741851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/18/2017] [Accepted: 04/30/2017] [Indexed: 11/18/2022] Open
Abstract
Many studies have been done to evaluate the effect of various natural products in controlling asthma symptoms. Virgin coconut oil (VCO) is known to contain active compounds that have beneficial effects on human health and diseases. The objective of this study was to evaluate the effect of VCO inhalation on airway remodelling in a rabbit model of allergic asthma. The effects of VCO inhalation on infiltration of airway inflammatory cells, airway structures, goblet cell hyperplasia, and cell proliferation following ovalbumin induction were evaluated. Allergic asthma was induced by a combination of ovalbumin and alum injection and/or followed by ovalbumin inhalation. The effect of VCO inhalation was then evaluated via the rescue or the preventive route. Percentage of inflammatory cells infiltration, thickness of epithelium and mucosa regions, and the numbers of goblet and proliferative cells were reduced in the rescue group but not in preventive group. Analysis using a gas chromatography-mass spectrometry found that lauric acid and capric acid were among the most abundant fatty acids present in the sample. Significant improvement was observed in rescue route in alleviating the asthma symptoms, which indicates the VCO was able to relieve asthma-related symptoms more than preventing the onset of asthma.
Collapse
Affiliation(s)
- N. A. Kamalaldin
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Penang, Malaysia
| | - S. A. Sulaiman
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - M. R. Yusop
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Penang, Malaysia
- School of Chemical Science and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - B. Yahaya
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Kepala Batas, Penang, Malaysia
| |
Collapse
|
34
|
Yu CJ, Cui XY, Lu L, Yang J, Chen B, Zhu CW, Gao X. Effects of glucocorticoid on the expression and regulation of aquaporin 5 in the paranasal sinus of rats with chronic rhinosinusitis. Exp Ther Med 2017; 13:1753-1756. [PMID: 28565763 PMCID: PMC5443270 DOI: 10.3892/etm.2017.4215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 12/23/2016] [Indexed: 01/11/2023] Open
Abstract
Aquaporins (AQPs) are water-specific membrane channel proteins that regulate water homeostasis for cells and organisms. AQP5 serves an important role in the maintenance of mucosal water homeostasis, and potentially contributes to mucosal edema and inflammation formation in chronic rhinosinusitis (CRS). The aim of the present study was to explore the expression pattern of AQP5 and the effect of glucocorticoids on AQP5 expression in rats with CRS. The rats were randomly divided into three equal groups, as follows: CRS, dexamethasone (dexa) treatment and control groups. A polyvinyl acetal material containing Staphylococcus aureus was inserted into the left nasal cavity of each rat from the CRS and dexa groups. On the 90th post-operative day, the dexa group received dexamethasone (2 mg/kg/day) via intraperitoneal injection for 7 days. The controls did not receive any treatment. The expression of AQP5 in the sinonasal mucosa was determined using immunohistochemistry and quantitative PCR. The immunoreactivities of AQP5 were primarily noted in the epithelial lining and glandular cells, the vascular endothelium and in the goblet cells in the sinonasal mucosa. The AQP5 mRNA expression level was significantly higher in the dexa group than in the control and CRS groups (P=0.006 and P=0.014, respectively). However, no significant difference was indicated between the CRS and control groups (P=0.760). In conclusion, the current study suggests that glucocorticoids induce AQP5 expression in the sinonasal mucosa of CRS rats, which highlights AQP5 as a potential target in the diagnosis and treatment of CRS.
Collapse
Affiliation(s)
- Chen-Jie Yu
- Department of Otolaryngology, Drum Tower Clinical Medical School, Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210008, P.R. China
| | - Xin-Yan Cui
- Department of Otolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ling Lu
- Department of Otolaryngology, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210008, P.R. China
| | - Jun Yang
- Department of Pathology, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210008, P.R. China
| | - Bin Chen
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Cheng-Wen Zhu
- Department of Otolaryngology, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210008, P.R. China
| | - Xia Gao
- Department of Otolaryngology, Drum Tower Clinical Medical School, Nanjing Medical University, Nanjing, Jiangsu 210008, P.R. China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
35
|
Wittekindt OH. Tight junctions in pulmonary epithelia during lung inflammation. Pflugers Arch 2017; 469:135-147. [PMID: 27921210 PMCID: PMC5203840 DOI: 10.1007/s00424-016-1917-3] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/31/2022]
Abstract
Inflammatory lung diseases like asthma bronchiale, chronic obstructive pulmonary disease and allergic airway inflammation are widespread public diseases that constitute an enormous burden to the health systems. Mainly classified as inflammatory diseases, the treatment focuses on strategies interfering with local inflammatory responses by the immune system. Inflammatory lung diseases predispose patients to severe lung failures like alveolar oedema, respiratory distress syndrome and acute lung injury. These life-threatening syndromes are caused by increased permeability of the alveolar and airway epithelium and exudate formation. However, the mechanism underlying epithelium barrier breakdown in the lung during inflammation is elusive. This review emphasises the role of the tight junction of the airway epithelium as the predominating structure conferring epithelial tightness and preventing exudate formation and the impact of inflammatory perturbations on their function.
Collapse
Affiliation(s)
- Oliver H Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
36
|
Fabregat G, García-de-la-Asunción J, Sarriá B, Mata M, Cortijo J, de Andrés J, Gallego L, Belda FJ. Expression of aquaporins 1 and 5 in a model of ventilator-induced lung injury and its relation to tidal volume. Exp Physiol 2016; 101:1418-1431. [PMID: 27424549 DOI: 10.1113/ep085729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 07/11/2016] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Although different studies have attempted to find factors that influence the expression of aquaporins (AQPs) in the lung in different situations, to date no research group has explored the expression of AQP1 and AQP5 jointly in rats mechanically ventilated with different tidal volumes in a model of ventilator-induced lung injury. What is the main finding? Mechanical ventilation with a high tidal volume causes lung injury and oedema, increasing lung permeability. In rats ventilated with a high tidal volume, the pulmonary expression of AQP1 decreases. We analysed the expression of aquaporins 1 and 5 and its relation with tidal volume in a model of ventilator-induced lung injury. Forty-two rats were used. Six non-ventilated animals were killed (control group). The remaining rats were ventilated for 2 h with different tidal volumes (group 7ML with 7 ml kg-1 and group 20ML with 20 ml kg-1 ) and a respiratory rate of 90 breaths min-1 . Lung oedema was measured, and the expression of AQP1 and AQP5 was determined by Western immunoblotting and measurement of mRNA. Lung oedema and alveolar-capillary membrane permeability were significantly increased in the animals of group 20ML compared with the control group. Expression of AQP1 was decreased in groups 7ML and 20ML compared with the control group. In conclusion, mechanical ventilation with a high tidal volume causes lung injury and oedema, increasing lung permeability. In rats ventilated with a high tidal volume, the pulmonary expression of AQP1 decreases.
Collapse
Affiliation(s)
| | | | | | - Manuel Mata
- School of Medicine, University of Valencia, Valencia, Spain
| | - Julio Cortijo
- School of Medicine, University of Valencia, Valencia, Spain
| | - José de Andrés
- Hospital General Universitario, Valencia, Spain.,School of Medicine, University of Valencia, Valencia, Spain
| | - Lucía Gallego
- Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Francisco Javier Belda
- Hospital Clinico Universitario, Valencia, Spain.,School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
37
|
Takeda K, Miyahara N, Matsubara S, Taube C, Kitamura K, Hirano A, Tanimoto M, Gelfand EW. Immunomodulatory Effects of Ambroxol on Airway Hyperresponsiveness and Inflammation. Immune Netw 2016; 16:165-75. [PMID: 27340385 PMCID: PMC4917400 DOI: 10.4110/in.2016.16.3.165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 02/07/2023] Open
Abstract
Ambroxol is used in COPD and asthma to increase mucociliary clearance and regulate surfactant levels, perhaps through anti-oxidant and anti-inflammatory activities. To determine the role and effect of ambroxol in an experimental model of asthma, BALB/c mice were sensitized to ovalbumin (OVA) followed by 3 days of challenge. Airway hyperresponsiveness (AHR), lung cell composition and histology, and cytokine and protein carbonyl levels in bronchoalveolar lavage (BAL) fluid were determined. Ambroxol was administered either before the first OVA challenge or was begun after the last allergen challenge. Cytokine production levels from lung mononuclear cells (Lung MNCs) or alveolar macrophages (AM) were also determined. Administration of ambroxol prior to challenge suppressed AHR, airway eosinophilia, goblet cell metaplasia, and reduced inflammation in subepithelial regions. When given after challenge, AHR was suppressed but without effects on eosinophil numbers. Levels of IL-5 and IL-13 in BAL fluid were decreased when the drug was given prior to challenge; when given after challenge, increased levels of IL-10 and IL-12 were detected. Decreased levels of protein carbonyls were detected in BAL fluid following ambroxol treatment after challenge. In vitro, ambroxol increased levels of IL-10, IFN-γ, and IL-12 from Lung MNCs and AM, whereas IL-4, IL-5, and IL-13 production was not altered. Taken together, ambroxol was effective in preventing AHR and airway inflammation through upregulation of Th1 cytokines and protection from oxidative stress in the airways.
Collapse
Affiliation(s)
- Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, U.S.A
| | - Nobuaki Miyahara
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, U.S.A
| | - Shigeki Matsubara
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, U.S.A
| | - Christian Taube
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, U.S.A
| | - Kenichi Kitamura
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Astushi Hirano
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Mitsune Tanimoto
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, U.S.A
| |
Collapse
|
38
|
Aquaporin-3 potentiates allergic airway inflammation in ovalbumin-induced murine asthma. Sci Rep 2016; 6:25781. [PMID: 27165276 PMCID: PMC4863152 DOI: 10.1038/srep25781] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/22/2016] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a pivotal role in the pathogenesis of asthma. Aquaporin-3 (AQP3) is a small transmembrane water/glycerol channel that may facilitate the membrane uptake of hydrogen peroxide (H2O2). Here we report that AQP3 potentiates ovalbumin (OVA)-induced murine asthma by mediating both chemokine production from alveolar macrophages and T cell trafficking. AQP3 deficient (AQP3(-/-)) mice exhibited significantly reduced airway inflammation compared to wild-type mice. Adoptive transfer experiments showed reduced airway eosinophilic inflammation in mice receiving OVA-sensitized splenocytes from AQP3(-/-) mice compared with wild-type mice after OVA challenge, consistently with fewer CD4(+) T cells from AQP3(-/-) mice migrating to the lung than from wild-type mice. Additionally, in vivo and vitro experiments indicated that AQP3 induced the production of some chemokines such as CCL24 and CCL22 through regulating the amount of cellular H2O2 in M2 polarized alveolar macrophages. These results imply a critical role of AQP3 in asthma, and AQP3 may be a novel therapeutic target.
Collapse
|
39
|
Wang L, Shan Y, Ye Y, Jin L, Zhuo Q, Xiong X, Zhao X, Lin L, Miao J. COX-2 inhibition attenuates lung injury induced by skeletal muscle ischemia reperfusion in rats. Int Immunopharmacol 2015; 31:116-22. [PMID: 26724476 DOI: 10.1016/j.intimp.2015.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/30/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Skeletal muscle ischemia reperfusion accounts for high morbidity and mortality, and cyclooxygenase (COX)-2 is implicated in causing muscle damage. Downregulation of aquaporin-1 (AQP-1) transmembrane protein is implicated in skeletal muscle ischemia reperfusion induced remote lung injury. The expression of COX-2 in lung tissue and the effect of COX-2 inhibition on AQP-1 expression and lung injury during skeletal muscle ischemia reperfusion are not known. We investigated the role of COX-2 in lung injury induced by skeletal muscle ischemia reperfusion in rats and evaluated the effects of NS-398, a specific COX-2 inhibitor. METHODS Twenty-four Sprague Dawley rats were randomized into 4 groups: sham group (SM group), sham+NS-398 group (SN group), ischemia reperfusion group (IR group) and ischemia reperfusion+NS-398 group (IN group). Rats in the IR and IN groups were subjected to 3h of bilateral ischemia followed by 6h of reperfusion in hindlimbs, and intravenous NS-398 8 mg/kg was administered in the IN group. In the SM and SN groups, rubber bands were in place without inflation. At the end of reperfusion, myeloperoxidase (MPO) activity, COX-2 and AQP-1 protein expression in lung tissue, PGE2 metabolite (PGEM), tumor necrosis factor (TNF)-α and interleukin (IL)-1β levels in bronchoalveolar lavage (BAL) fluid were assessed. Histological changes in lung and muscle tissues and wet/dry (W/D) ratio were also evaluated. RESULTS MPO activity, COX-2 expression, W/D ratio in lung tissue, and PGEM, TNF-α and IL-1β levels in BAL fluid were significantly increased, while AQP-1 protein expression downregulated in the IR group as compared to that in the SM group (P<0.05). These changes were remarkably mitigated in the IN group (P<0.05). NS-398 treatment also alleviated histological signs of lung and skeletal muscle injury. CONCLUSION COX-2 protein expression was upregulated in lung tissue in response to skeletal muscle ischemia reperfusion. COX-2 inhibition may modulate pulmonary AQP-1 expression and attenuate lung injury.
Collapse
Affiliation(s)
- Liangrong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Yuanlu Shan
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Yuzhu Ye
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Lida Jin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Qian Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Xiangqing Xiong
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Xiyue Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - Lina Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| | - JianXia Miao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, People's Republic of China.
| |
Collapse
|
40
|
Aquaporin-1 Deficiency Protects Against Myocardial Infarction by Reducing Both Edema and Apoptosis in Mice. Sci Rep 2015; 5:13807. [PMID: 26348407 PMCID: PMC4562302 DOI: 10.1038/srep13807] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/06/2015] [Indexed: 01/26/2023] Open
Abstract
Many studies have determined that AQP1 plays an important role in edema formation and resolution in various tissues via water transport across the cell membrane. The aim of this research was to determine both if and how AQP1 is associated with cardiac ischemic injury, particularly the development of edema following myocardial infarction (MI). AQP1+/+ and AQP1−/− mice were used to create the MI model. Under physiological conditions, AQP1−/− mice develop normally; however, in the setting of MI, they exhibit cardioprotective properties, as shown by reduced cardiac infarct size determined via NBT staining, improved cardiac function determined via left ventricular catheter measurements, decreased AQP1-dependent myocardial edema determined via water content assays, and decreased apoptosis determined via TUNEL analysis. Cardiac ischemia caused by hypoxia secondary to AQP1 deficiency stabilized the expression of HIF-1α in endothelial cells and subsequently decreased microvascular permeability, resulting in the development of edema. The AQP1-dependent myocardial edema and apoptosis contributed to the development of MI. AQP1 deficiency protected cardiac function from ischemic injury following MI. Furthermore, AQP1 deficiency reduced microvascular permeability via the stabilization of HIF-1α levels in endothelial cells and decreased cellular apoptosis following MI.
Collapse
|
41
|
Han G, Ma L, Guo Y, Li L, Li D, Liu H. Hyperbaric oxygen therapy palliates lipopolysaccharide-induced acute lung injury in rats by upregulating AQP1 and AQP5 expression. Exp Lung Res 2015; 41:444-9. [PMID: 26317897 DOI: 10.3109/01902148.2015.1064189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE Hyperbaric oxygen (HBO) therapy has been suggested to palliate acute lung injury (ALI), but the mechanisms involved are not well understood. This study is to elucidate the involvement of AQP1 and AQP5 in the HBO related ALI therapy. MATERIALS AND METHODS lipopolysaccharide (LPS) was administrated into SD rats to obtain ALI models. Pressure of oxygen (PaO2) and carbon dioxide (PaCO2) in arterial blood and oxygenation index in rats after LPS and HBO treatments were determined. Pathological changes of the lungs were examined by hematoxylin and eosin staining. Alteration of TNF-α level during LPS and HBO treatments was evaluated with ELISA analysis. Western blot was employed to assess the expression of AQP1 and AQP5. RESULTS Blood gas indexes were largely decreased by LPS administration, which responded to HBO. Pathological examination showed that the inflammation symptoms in lungs induced by LPS were also palliated after HBO preconditioning. LPS induced the expression of TNF-α at a high level which could be downregulated by HBO and TNF-α antagonist treatments. Results of AQP1 and AQP5 determination found that HBO and TNF-α antagonist would upregulate the expression of AQP1 and AQP5 which was inhibited in rats with ALI. CONCLUSIONS HBO therapy palliated LPS-induced ALI in rats by downregulating TNF-α expression. HBO also upregulated AQP1 and AQP5 expression. These results could serve as guidelines for the full understanding of ALI therapy by HBO, thus achieving maximized therapeutic efficiency.
Collapse
Affiliation(s)
- Guang Han
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| | - Ling Ma
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| | - Yao Guo
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| | - Lu Li
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| | - Dan Li
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| | - Hongtao Liu
- a Department of Anesthesia , Shengjing Hospital of China Medical University , Shenyang , Liaoning , China
| |
Collapse
|
42
|
Gregoire F, Lucidi V, Zerrad-Saadi A, Virreira M, Bolaky N, Delforge V, Lemmers A, Donckier V, Devière J, Demetter P, Perret J, Delporte C. Analysis of aquaporin expression in liver with a focus on hepatocytes. Histochem Cell Biol 2015; 144:347-63. [PMID: 26126651 DOI: 10.1007/s00418-015-1341-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2015] [Indexed: 12/30/2022]
Abstract
A deeper understanding of aquaporins (AQPs) expression and transcriptional regulation will provide useful information for liver pathophysiology. We established a complete AQPs mRNA expression profile in human and mouse liver, as well as protein localization of expressed AQPs. Additionally, the modulation of AQPs mRNA levels in response to various agents was determined in human HuH7 cells and in primary culture of mouse hepatocytes. AQP1, AQP3, AQP7, AQP8, and AQP9 mRNA and protein expressions were detected in human liver, while only AQP6 and AQP11 mRNAs were detected. We reported for the first time the localization of AQP3 in Kupffer cells, AQP7 in hepatocytes and endothelial cells, and AQP9 in cholangiocytes. In addition, we confirmed the localization of AQP1 in endothelial cells, and of AQP8 and AQP9 in hepatocytes. On HuH7 cells, we reported the presence of AQP4 mRNA, confirmed the presence of AQP3, AQP7, and AQP11 mRNAs, but not of AQP8 mRNA. On primary culture of murine hepatocytes, AQP1 and AQP7 mRNAs were identified, while the presence of AQP3, AQP8, AQP9, and AQP11 mRNAs was confirmed. At the protein level, murine endothelial liver cells expressed AQP1 and AQP9, while hepatocytes expressed AQP3, AQP7, AQP8, and AQP9, and macrophages expressed AQP3. Dexamethasone, forskolin, AICAR, rosiglitazone, octanoylated, and non-octanoylated ghrelin regulated some AQP expression in primary culture of murine hepatocytes and human HuH7 cells. Additional studies will be required to further assess the role of AQPs expression in human and murine liver and understand the transcriptional regulation of AQPs in hepatocytes under pathophysiological conditions.
Collapse
Affiliation(s)
- Françoise Gregoire
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Valério Lucidi
- Digestive Oncology Department, Erasme Hospital, Brussels, Belgium
| | - Amal Zerrad-Saadi
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Myrna Virreira
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Nargis Bolaky
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Valérie Delforge
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Arnaud Lemmers
- Gastroenterology Department, Erasme Hospital, Brussels, Belgium
| | - Vincent Donckier
- Digestive Oncology Department, Erasme Hospital, Brussels, Belgium
| | - Jacques Devière
- Gastroenterology Department, Erasme Hospital, Brussels, Belgium
| | - Pieter Demetter
- Anatomopathology Department, Erasme Hospital, Brussels, Belgium
| | - Jason Perret
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
43
|
miR-96 and miR-330 overexpressed and targeted AQP5 in lipopolysaccharide-induced rat lung damage of disseminated intravascular coagulation. Blood Coagul Fibrinolysis 2015; 25:731-7. [PMID: 24806323 DOI: 10.1097/mbc.0000000000000133] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Disseminated intravascular coagulation (DIC) is a severe clinical condition that can lead to or aggravate the development of multiple organ dysfunction syndrome. Of all types of organ damage, lung damage is the most frequent and most severe. In DIC patients, lung damage is primarily characterized by pulmonary edema. Aquaporin (AQP) 5 is the chief AQP in the lungs and it plays a key role in many processes, including water transport in normal and abnormal lungs. Here we demonstrate that expression of AQP5 and two microRNAs, miR-96 and miR-330, in rat lung of lipopolysaccharide (LPS)-induced DIC. We also show that both miR-96 and miR-330 can regulate the expression of AQP5 by binding with its 3'-untranslated region (UTR) by luciferase activity assay. These results suggest that microRNAs are involved in lung damage in LPS-induced rat DIC and can be a potential target for molecular therapy.
Collapse
|
44
|
Wang W, Wang X, Ma L, Zhang R. Histamine downregulates aquaporin 5 in human nasal epithelial cells. Am J Rhinol Allergy 2015; 29:188-92. [PMID: 25781725 DOI: 10.2500/ajra.2015.29.4181] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Aquaporin 5 (AQP5) is a water-specific channel protein. It is thought to be a key participant in fluid secretion and a rate-limiting barrier to the secretion seen during allergic inflammation. We sought to determine the effect of histamine on AQP5 expression in human nasal epithelial cells (HNEpC). METHODS HNEpC cells were cultured with four concentrations of histamine in vitro. The phosphorylation of cyclic adenosine monophosphate-responsive element binding protein (CREB) at serine 133 and the AQP5 protein were measured by using immunocytochemistry and Western blotting. Real-time polymerase chain reaction was used to detect AQP5 messenger ribonucleic acid (mRNA). RESULTS Concentration-dependent histamine induced-inhibition of CREB phosphorylation at serine 133 in HNEpC cells was observed, and AQP5 mRNA and protein were also downregulated in a concentration-dependent fashion. CONCLUSION Histamine downregulates AQP5 production in HNEpC cells by inhibiting CREB phosphorylation at serine 133.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Anatomy, Schools of Medicine and Nursing Sciences, Huzhou University, China
| | | | | | | |
Collapse
|
45
|
Aquaporin 5 expression in mouse mammary gland cells is not driven by promoter methylation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:460598. [PMID: 25767807 PMCID: PMC4342075 DOI: 10.1155/2015/460598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/16/2015] [Indexed: 11/17/2022]
Abstract
Several studies have revealed that aquaporins play a role in tumor progression and invasion. In breast carcinomas, high levels of aquaporin 5 (AQP5), a membrane protein involved in water transport, have been linked to increased cell proliferation and migration, thus facilitating tumor progression. Despite the potential role of AQP5 in mammary oncogenesis, the mechanisms controlling mammary AQP5 expression are poorly understood. In other tissues, AQP5 expression has been correlated with its promoter methylation, yet, very little is known about AQP5 promoter methylation in the mammary gland. In this work, we used the mouse mammary gland cell line EpH4, in which we controlled AQP5 expression via the steroid hormone dexamethasone (Dex) to further investigate mechanisms regulating AQP5 expression. In this system, we observed a rapid drop of AQP5 mRNA levels with a delay of several hours in AQP5 protein, suggesting transcriptional control of AQP5 levels. Yet, AQP5 expression was independent of its promoter methylation, or to the presence of negative glucocorticoid receptor elements (nGREs) in its imminent promoter region, but was rather influenced by the cell proliferative state or cell density. We conclude that AQP5 promoter methylation is not a universal mechanism for AQP5 regulation and varies on cell and tissue type.
Collapse
|
46
|
Increased expression of AQP 1 and AQP 5 in rat lungs ventilated with low tidal volume is time dependent. PLoS One 2014; 9:e114247. [PMID: 25489856 PMCID: PMC4260863 DOI: 10.1371/journal.pone.0114247] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/05/2014] [Indexed: 01/24/2023] Open
Abstract
Background and Goals Mechanical ventilation (MV) can induce or worsen pulmonary oedema. Aquaporins (AQPs) facilitate the selective and rapid bi-directional movement of water. Their role in the development and resolution of pulmonary oedema is controversial. Our objectives are to determine if prolonged MV causes lung oedema and changes in the expression of AQP 1 and AQP 5 in rats. Methods 25 male Wistar rats were subjected to MV with a tidal volume of 10 ml/kg, during 2 hours (n = 12) and 4 hours (n = 13). Degree of oedema was compared with a group of non-ventilated rats (n = 5). The expression of AQP 1 and AQP 5 were determined by western immunoblotting, measuring the amount of mRNA (previously amplified by RT-PCR) and immunohistochemical staining of AQPs 1 and 5 in lung samples from all groups. Results Lung oedema and alveolar-capillary membrane permeability did not change during MV. AQP-5 steady state levels in the western blot were increased (p<0.01) at 2 h and 4 h of MV. But in AQP-1 expression these differences were not found. However, the amount of mRNA for AQP-1 was increased at 2 h and 4 h of MV; and for AQP 5 at 4 h of MV. These findings were corroborated by representative immunohistochemical lung samples. Conclusion In lungs from rats ventilated with a low tidal volume the expression of AQP 5 increases gradually with MV duration, but does not cause pulmonary oedema or changes in lung permeability. AQPs may have a protective effect against the oedema induced by MV.
Collapse
|
47
|
Calero C, López-Campos JL, Izquierdo LG, Sánchez-Silva R, López-Villalobos JL, Sáenz-Coronilla FJ, Arellano-Orden E, Montes-Worboys A, Echevarría M. Expression of aquaporins in bronchial tissue and lung parenchyma of patients with chronic obstructive pulmonary disease. Multidiscip Respir Med 2014; 9:29. [PMID: 24917931 PMCID: PMC4050095 DOI: 10.1186/2049-6958-9-29] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/29/2014] [Indexed: 11/10/2022] Open
Abstract
Background Aquaporins AQP1 and AQP5 are highly expressed in the lung. Recent studies have shown that the expression of these proteins may be mechanistically involved in the airway inflammation and in the pathogenesis of chronic obstructive pulmonary disease (COPD). The aim of this study was to investigate the expression of AQP1 and AQP5 in the bronchial tissue and the lung parenchyma of patients with COPD and COPD-resistant smokers. Methods Using a case–control design, we selected a group of 15 subjects with COPD and 15 resistant smokers (smokers without COPD) as a control, all of whom were undergoing lung resection surgery due to a lung neoplasm. We studied the expression of AQP1 and AQP5 in the bronchial tissue and the lung parenchyma by means of immunohistochemistry and reverse-transcription real-time polymerase chain reaction. Tissue expression of AQP1 and AQP5 was semi-quantitatively assessed in terms of intensity and expression by immunohistochemistry using a 4-point scale ranging from 0 (none) to 3 (maximum). Results There were no significant differences in gene expression between COPD patients and resistant smokers both in the bronchial tissue and in the lung parenchyma. However, AQP1 gene expression was 2.41-fold higher in the parenchyma of smokers with COPD compared to controls, whereas the AQP5 gene showed the opposite pattern, with a 7.75-fold higher expression in the bronchus of smokers with COPD compared with controls. AQP1 and AQP5 proteins were preferentially expressed in endothelial cells, showing a higher intensity for AQP1 (66.7% of cases with an intensity of 3, and 93.3% of subjects with an extension of 3 among patients with COPD). Subtle interstitial disease was associated with type II pneumocyte hyperplasia and an increased expression of AQP1. Conclusions This study provides pilot observations on the differences in AQP1 and AQP5 expression between COPD patients and COPD-resistant smokers. Our findings suggest a potential role for AQP1 in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Carmen Calero
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocio, Sevilla, Spain ; Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain ; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Luis López-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocio, Sevilla, Spain ; Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain ; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Lourdes Gómez Izquierdo
- Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Rocío Sánchez-Silva
- Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain
| | - Jose Luis López-Villalobos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocio, Sevilla, Spain ; Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain
| | | | - Elena Arellano-Orden
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocio, Sevilla, Spain ; Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain
| | - Ana Montes-Worboys
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocio, Sevilla, Spain ; Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain ; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Echevarría
- Instituto de Biomedicina de Sevilla (IBiS), Avda. Manuel Siurot, s/n 41013, Sevilla, Spain ; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
48
|
TNF-α regulates natriuretic peptides and aquaporins in human bronchial epithelial cells BEAS-2B. Mediators Inflamm 2013; 2013:159349. [PMID: 24369440 PMCID: PMC3863520 DOI: 10.1155/2013/159349] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/26/2013] [Accepted: 10/07/2013] [Indexed: 02/03/2023] Open
Abstract
Postoperative-fluid retention is a severe complication frequently reported in patients undergoing major surgical procedures. The complex network of molecules involved in such a severe surgery-induced condition remains poorly understood. Inflammation has been proposed among the various causes of fluid retention. Since TNF-α is one of the main proinflammatory cytokine initially released after major surgery, it is reasonable to assume its involvement in fluid overload. Here, we showed that TNF-α selectively regulates key molecules involved in fluids balance, such as natriuretic peptides (NPs) and aquaporins, in human bronchial epithelial cells BEAS-2B. In particular, we found that TNF-α induced a decrease of arial natriuretic peptide, natriuretic peptide receptor-1, aquaporin-1 and aquaporin-5 and an increase of brain natriuretic peptide with a different involvement of nuclear factor-κB and mitogen-activated protein kinases signaling pathway activation. Moreover, the observed changes in NPs expression, demonstrate inflammation as an additional cause of brain natriuretic peptide elevation, adding an important piece of information in the novel area of study regarding NPs and inflammation. Finally, we suggest that inflammation is one of the mechanisms of Aquaporin-1 and aquaporin-5 expression regulation. Therefore, in this exploratory study, we speculate that TNF-α might be involved in postoperative-fluid retention related to major surgery.
Collapse
|
49
|
She J, Bi J, Tong L, Song Y, Bai C. New insights of aquaporin 5 in the pathogenesis of high altitude pulmonary edema. Diagn Pathol 2013; 8:193. [PMID: 24274330 PMCID: PMC3937216 DOI: 10.1186/1746-1596-8-193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/26/2013] [Indexed: 11/28/2022] Open
Abstract
Background High altitude pulmonary edema (HAPE) affects individuals and is characterized by alveolar flooding with protein-rich edema as a consequence of blood-gas barrier disruption. In this study, we hypothesized that aquaporin 5 (AQP5) which is one kind of water channels may play a role in preservation of alveolar epithelial barrier integrity in high altitude pulmonary edema (HAPE). Methods Therefore, we established a model in Wildtype mice and AQP5 −/− mice were assingned to normoxic rest (NR), hypoxic rest (HR) and hypoxic exercise (HE) group. Mice were produced by training to walk at treadmill for exercising and chamber pressure was reduced to simulate climbing an altitude of 5000 m for 48 hours. Studies using BAL in HAPE mice to demonstrated that edema is caused leakage of albumin proteins and red cells across the alveolarcapillary barrier in the absence of any evidence of inflammation. Results In this study, the Lung wet/dry weight ratio and broncholalveolar lavage protein concentrations were slightly increased in HE AQP5 −/− mice compared to wildtype mice. And histologic evidence of hemorrhagic pulmonary edema was distinctly shown in HE group. The lung Evan’s blue permeability of HE group was showed slightly increased compare to the wildtype groups, and HR group was showed a medium situation from normal to HAPE development compared with NR and HE group. Conclusions Deletion of AQP5 slightly increased lung edema and lung injury compared to wildtype mice during HAPE development, which suggested that the AQP5 plays an important role in HAPE formation induced by high altitude simulation.
Collapse
Affiliation(s)
| | | | | | | | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, NO,180 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
50
|
Cataldi M, Sblendorio V, Leo A, Piazza O. Biofilm-dependent airway infections: a role for ambroxol? Pulm Pharmacol Ther 2013; 28:98-108. [PMID: 24252805 DOI: 10.1016/j.pupt.2013.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/31/2013] [Accepted: 11/11/2013] [Indexed: 11/16/2022]
Abstract
Biofilms are a key factor in the development of both acute and chronic airway infections. Their relevance is well established in ventilator associated pneumonia, one of the most severe complications in critically ill patients, and in cystic fibrosis, the most common lethal genetic disease in Caucasians. Accumulating evidence suggests that biofilms could have also a role in chronic obstructive pulmonary disease and their involvement in bronchiectasis has been proposed as well. When they grow in biofilms, microorganisms become multidrug-resistant. Therefore the treatment of biofilm-dependent airway infections is problematic. Indeed, it still largely based on measures aiming to prevent the formation of biofilms or remove them once that they are formed. Here we review recent evidence suggesting that the mucokinetic drug ambroxol has specific anti-biofilm properties. We also discuss how additional pharmacological properties of this drug could be beneficial in biofilm-dependent airway infections. Specifically, we review the evidence showing that: 1-ambroxol exerts anti-inflammatory effects by inhibiting at multiple levels the activity of neutrophils, and 2-it improves mucociliary clearance by interfering with the activity of airway epithelium ion channels and transporters including sodium/bicarbonate and sodium/potassium/chloride cotransporters, cystic fibrosis transmembrane conductance regulator and aquaporins. As a whole, the data that we review here suggest that ambroxol could be helpful in biofilm-dependent airway infections. However, considering the limited clinical evidence available up to date, further clinical studies are required to support the use of ambroxol in these diseases.
Collapse
Affiliation(s)
- M Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Via Pansini 5, 80131 Napoli, Italy.
| | - V Sblendorio
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Federico II University of Naples, Via Pansini 5, 80131 Napoli, Italy
| | - A Leo
- Department of Health Sciences, University Magna Græcia of Catanzaro, University Campus "Salvatore Venuta", Viale Europa, I-88100 Catanzaro, Italy
| | - O Piazza
- University of Salerno, Via Allende, 84081 Baronissi, Italy
| |
Collapse
|