1
|
de Souza ABF, de Matos NA, Castro TDF, Costa GDP, Talvani A, Nagato AC, de Menezes RCA, Bezerra FS. Preventive effects of hesperidin in an experimental model ofs acute lung inflammation. Respir Physiol Neurobiol 2024; 323:104240. [PMID: 38417564 DOI: 10.1016/j.resp.2024.104240] [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: 01/03/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024]
Abstract
In this study, we hypothesized that long-term administration of hesperidin can modulate the inflammatory response and oxidative stress in animals submitted to mechanical ventilation (MV). Twenty-five C57BL/6 male mice were divided into 5 groups: control, MV, animals receiving hesperidin in three doses 10, 25 and 50 mg/kg. The animals received the doses of hesperidin for 30 days via orogastric gavage, and at the end of the period the animals were submitted to MV. In animals submitted to MV, increased lymphocyte, neutrophil and monocyte/macrophage cell counts were observed in the blood and airways. Associated to this, MV promoted an increase in inflammatory cytokine levels such as CCL2, IL-12 and TNFα. The daily administration of hesperidin in the three doses prevented the effects caused by MV, which was observed by a lower influx of inflammatory cells into the airways, a reduction in inflammatory markers and less oxidative damage.
Collapse
Affiliation(s)
- Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Akinori Cardozo Nagato
- Immunopathology Laboratory and Experimental Pathology, Reproductive Biology Center (CRB), Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Rodrigo Cunha Alvim de Menezes
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Caputo LDS, Alves CDL, Laranjeira IM, Fonseca-Rodrigues D, da Silva Filho AA, Dias ACP, Pinto-Ribeiro F, Pereira Junior ODS, de Paula ACC, Nagato AC, Corrêa JODA. Copaiba oil minimizes inflammation and promotes parenchyma re-epithelization in acute allergic asthma model induced by ovalbumin in BALB/c mice. Front Pharmacol 2024; 15:1356598. [PMID: 38666018 PMCID: PMC11043548 DOI: 10.3389/fphar.2024.1356598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction: Asthma is a condition of airflow limitation, common throughout the world, with high mortality rates, especially as it still faces some obstacles in its management. As it constitutes a public health challenge, this study aimed to investigate the effect of copaiba oil (e.g., Copaifera langsdorffii), as a treatment resource, at doses of 50 and 100 mg/kg on certain mediators of acute lung inflammation (IL-33, GATA3, FOXP3, STAT3, and TBET) and early mechanisms of lung remodeling (degradation of elastic fiber tissues, collagen deposition, and goblet cell hyperplasia). Methods: Using an ovalbumin-induced acute allergic asthma model in BALB/c mice, we analyzed the inflammatory mediators through immunohistochemistry and the mechanisms of lung remodeling through histopathology, employing orcein, Masson's trichrome, and periodic acid-Schiff staining. Results: Copaiba oil treatment (CO) reduced IL-33 and increased FOXP3 by stimulating the FOXP3/GATA3 and FOXP3/STAT3 pathways. Additionally, it upregulated TBET, suggesting an additional role in controlling GATA3 activity. In the respiratory epithelium, CO decreased the fragmentation of elastic fibers while increasing the deposition of collagen fibers, favoring epithelial restructuring. Simultaneously, CO reduced goblet cell hyperplasia. Discussion: Although additional research is warranted, the demonstrated anti-inflammatory and re-epithelializing action makes CO a viable option in exploring new treatments for acute allergic asthma.
Collapse
Affiliation(s)
- Ludmila de Souza Caputo
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina de Lima Alves
- Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Inês Martins Laranjeira
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Centre of Molecular and Environmental Biology, CBMA, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Diana Fonseca-Rodrigues
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
| | | | - Alberto Carlos Pires Dias
- Centre of Molecular and Environmental Biology, CBMA, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Filipa Pinto-Ribeiro
- Life and Health Sciences Research Institute, ICVS, School of Medicine, Campus of Gualtar, University of Minho, Braga, Portugal
- ICVS/3B‟s - PT Government Associate Laboratory, Braga, Portugal
| | | | | | - Akinori Cardozo Nagato
- Department of Physiology, Federal University of Juiz de Fora, UFJF, Juiz de Fora, Brazil
| | | |
Collapse
|
3
|
Tenfen L, Simon Machado R, Mathias K, Piacentini N, Joaquim L, Bonfante S, Danielski LG, Engel NA, da Silva MR, Rezin GT, de Quadros RW, Gava FF, Petronilho F. Short-term hyperoxia induced mitochondrial respiratory chain complexes dysfunction and oxidative stress in lung of rats. Inhal Toxicol 2024; 36:174-188. [PMID: 38449063 DOI: 10.1080/08958378.2024.2322497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 02/18/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Oxygen therapy is an alternative for many patients with hypoxemia. However, this practice can be dangerous as oxygen is closely associated with the development of oxidative stress. METHODS Male Wistar rats were exposed to hyperoxia with a 40% fraction of inspired oxygen (FIO2) and hyperoxia (FIO2 = 60%) for 120 min. Blood and lung tissue samples were collected for gas, oxidative stress, and inflammatory analyses. RESULTS Hyperoxia (FIO2 = 60%) increased PaCO2 and PaO2, decreased blood pH and caused thrombocytopenia and lymphocytosis. In lung tissue, neutrophil infiltration, nitric oxide concentration, carbonyl protein formation and the activity of complexes I and II of the mitochondrial respiratory chain increased. FIO2 = 60% decreased SOD activity and caused several histologic changes. CONCLUSION In conclusion, we have experimentally demonstrated that short-term exposure to high FIO2 can cause oxidative stress in the lung.
Collapse
Affiliation(s)
- Leonardo Tenfen
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Richard Simon Machado
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Khiany Mathias
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Natalia Piacentini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Larissa Joaquim
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Sandra Bonfante
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Lucineia Gainski Danielski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Nicole Alessandra Engel
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Mariella Reinol da Silva
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | - Gislaine Tezza Rezin
- Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, Brazil
| | | | - Fernanda Frederico Gava
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
4
|
Li J, Bao T, Cao L, Ma M, Yu B, Zhang Y, Wu R, Zhu H, Tian Z. Establishment of a juvenile mouse asthma model induced by postnatal hyperoxia exposure combined with early OVA sensitization. Heliyon 2024; 10:e23291. [PMID: 38148813 PMCID: PMC10750071 DOI: 10.1016/j.heliyon.2023.e23291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Objective To establish a juvenile mouse asthma model by postnatal hyperoxia exposure combined with early ovalbumin (OVA) sensitization. Methods Female C57BL/6J newborn mice were exposed to hyperoxia (95 % O2) from postnatal day-1 (PND1) to PND7; intraperitoneally injected with OVA suspension on PND21, PND28; and stimulated by nebulized inhalation of 1 % OVA from PND36 to PND42. Within 48 h of the last challenge, we observed their activity performance and evaluated airway responsiveness (AHR). All mice were executed at PND44. Female (n = 32) were divided into four groups as follows: room air(RA)+phosphate-buffered saline (PBS) group; O2 (hyperoxia, 95 % O2) + PBS group; RA + OVA group; O2+OVA group. We obtained the serum, bronchoalveolar lavage fluid (BALF), and lung tissues. The Wright-Giemsa staining was performed for leukocyte classification in BALF and HE staining for pathological examination. The levels of IL-2, IL-5, IL-13, IL-17A and IL-10 in BALF and tIgE and sIgE in serum were detected by ELISA. Results Compared with OVA sensitization or hyperoxia exposure alone, the mice in the model group (O2+OVA) showed asthma-like symptoms and increased airway hyperreactivity,The levels of IL-5,IL-13 IL-17A were increased in BLAF,and total leukocyte and eosinophil counts were also significant increasesed. The levels of tIgE and sIgE in serum were increased. Conclusion Postnatal hyperoxia exposure combined with early OVA sensitization might establish a juvenile mouse asthma model.
Collapse
Affiliation(s)
- Jingyan Li
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Linxia Cao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Bingrui Yu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Yuan Zhang
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Rong Wu
- Neonatal Medical Center, Huaian Maternity and Child Healthcare Hospital, Anhui Medical University, Huai'an, Jiangsu, 223002, China
| | - Haiyan Zhu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| |
Collapse
|
5
|
Li J, Bao T, Cao L, Ma M, Zhang Y, Tian Z. Effects of early postnatal hyperoxia exposure combined with early ovalbumin sensitization on lung inflammation and bacterial flora in a juvenile mouse model of asthma. Front Microbiol 2023; 14:1220042. [PMID: 37485534 PMCID: PMC10358775 DOI: 10.3389/fmicb.2023.1220042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Objective The aim of this study is to explore the effects of early postnatal hyperoxia exposure combined with early ovalbumin (OVA) sensitization on lung inflammation and bacterial flora in neonatal mice on a juvenile mouse model of asthma. Methods Thirty-two newborn female C57BL/6 J mice were randomly divided into four groups, which including room air+phosphate-buffered saline (PBS) group, hyperoxia+PBS group, room air+OVA group, and hyperoxia+OVA group, according to the hyperoxia exposure and/or OVA induction. Mice were exposed to either 95% O2 or room air for 7 days after birth; after 7 days, they were exposed to air and received an intraperitoneal injection of OVA suspension or PBS solution on postnatal days 21 (P21) and 28 (P28). From P36 to P42, the mice were allowed to inhale of 1% OVA or 0.9% NaCl solution. The mice were observed after the last excitation. HE staining was performed to observe the pathological changes in lung tissues. Wright-Giemsa staining was used to perform bronchoalveolar lavage fluid (BALF) leukocyte sorting. Enzyme-linked immunosorbent assay was used to determined the cytokines levels of interleukin (IL)-2, IL-5, IL-13, IL-17A, and IL-10 and serum IgE levels in BALF. Additionally, 16S rRNA sequencing was used to analyze the characteristics of lung microbiota. Results Mice in the hyperoxia+OVA group showed asthma-like symptoms. HE staining results revealed a significant thickening of the airway wall and airway inflammation. BALF analysis of cellular components showed significant increases in total leukocyte and eosinophil counts and the levels of cytokines related to Th2 (IL-5 and IL-13) and Th17 (IL-17A); 16S rRNA sequencing revealed that the main members of the pulmonary microflora were Actinobacteriota, Proteobacteria, Firmicutes, and Bacteroidota at the phylum level. In addition, the bacteria with a major role were Acinetobacter and Moraxellaceae in the O2 + OVA group. Conclusion The mouse suffering from postnatal hyperoxia exposure and early OVA sensitization, changes in symptoms, pathology, leukocyte and eosinophil counts, and levels of different T-cell cytokines in BALF and lung microbiota, which may provide a basis for the establishment of a juvenile mouse model of asthma.
Collapse
|
6
|
Hirani D, Alvira CM, Danopoulos S, Milla C, Donato M, Tian L, Mohr J, Dinger K, Vohlen C, Selle J, Koningsbruggen-Rietschel SV, Barbarino V, Pallasch C, Rose-John S, Odenthal M, Pryhuber GS, Mansouri S, Savai R, Seeger W, Khatri P, Al Alam D, Dötsch J, Alejandre Alcazar MA. Macrophage-derived IL-6 trans-signaling as a novel target in the pathogenesis of bronchopulmonary dysplasia. Eur Respir J 2021; 59:13993003.02248-2020. [PMID: 34446466 PMCID: PMC8850688 DOI: 10.1183/13993003.02248-2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
Rationale Premature infants exposed to oxygen are at risk for bronchopulmonary dysplasia (BPD), which is characterised by lung growth arrest. Inflammation is important, but the mechanisms remain elusive. Here, we investigated inflammatory pathways and therapeutic targets in severe clinical and experimental BPD. Methods and results First, transcriptomic analysis with in silico cellular deconvolution identified a lung-intrinsic M1-like-driven cytokine pattern in newborn mice after hyperoxia. These findings were confirmed by gene expression of macrophage-regulating chemokines (Ccl2, Ccl7, Cxcl5) and markers (Il6, Il17A, Mmp12). Secondly, hyperoxia-activated interleukin 6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) signalling was measured in vivo and related to loss of alveolar epithelial type II cells (ATII) as well as increased mesenchymal marker. Il6 null mice exhibited preserved ATII survival, reduced myofibroblasts and improved elastic fibre assembly, thus enabling lung growth and protecting lung function. Pharmacological inhibition of global IL-6 signalling and IL-6 trans-signalling promoted alveolarisation and ATII survival after hyperoxia. Third, hyperoxia triggered M1-like polarisation, possibly via Krüppel-like factor 4; hyperoxia-conditioned medium of macrophages and IL-6-impaired ATII proliferation. Finally, clinical data demonstrated elevated macrophage-related plasma cytokines as potential biomarkers that identify infants receiving oxygen at increased risk of developing BPD. Moreover, macrophage-derived IL6 and active STAT3 were related to loss of epithelial cells in BPD lungs. Conclusion We present a novel IL-6-mediated mechanism by which hyperoxia activates macrophages in immature lungs, impairs ATII homeostasis and disrupts elastic fibre formation, thereby inhibiting lung growth. The data provide evidence that IL-6 trans-signalling could offer an innovative pharmacological target to enable lung growth in severe neonatal chronic lung disease. M1-like macrophage activation is linked to IL-6/STAT3 axis in clinical and experimental BPD. Inhibition of macrophage-related IL-6 trans-signalling promotes ATII survival and lung growth in experimental BPD as a new therapy for preterm infants.https://bit.ly/3AhF7GP
Collapse
Affiliation(s)
- Dharmesh Hirani
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Carlos Milla
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michele Donato
- Biomedical Informatics Research-Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, USA
| | - Jasmine Mohr
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Katharina Dinger
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Christina Vohlen
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany
| | - Silke V Koningsbruggen-Rietschel
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Verena Barbarino
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, University of Cologne, Koln, Germany
| | - Christian Pallasch
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, University of Cologne, Koln, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Margarete Odenthal
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute for Pathology, Koln, Germany
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Siavash Mansouri
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL)
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL)
| | - Purvesh Khatri
- Biomedical Informatics Research-Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California, USA
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Miguel A Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany .,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL).,University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
7
|
Sidramagowda Patil S, Hernández-Cuervo H, Fukumoto J, Krishnamurthy S, Lin M, Alleyn M, Breitzig M, Narala VR, Soundararajan R, Lockey RF, Kolliputi N, Galam L. Alda-1 Attenuates Hyperoxia-Induced Acute Lung Injury in Mice. Front Pharmacol 2021; 11:597942. [PMID: 33597876 PMCID: PMC7883597 DOI: 10.3389/fphar.2020.597942] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Acute lung injury (ALI), a milder form of acute respiratory distress syndrome (ARDS), is a leading cause of mortality in older adults with an increasing prevalence. Oxygen therapy, is a common treatment for ALI, involving exposure to a high concentration of oxygen. Unfortunately, hyperoxia induces the formation of reactive oxygen species which can cause an increase in 4-HNE (4-hydroxy 2 nonenal), a toxic byproduct of lipid peroxidation. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) serves as an endogenous shield against oxidative stress-mediated damage by clearing 4-HNE. Alda-1 [(N-(1, 3 benzodioxol-5-ylmethyl)-2, 6- dichloro-benzamide)], a small molecular activator of ALDH2, protects against reactive oxygen species-mediated oxidative stress by promoting ALDH2 activity. As a result, Alda-1 shields against ischemic reperfusion injury, heart failure, stroke, and myocardial infarction. However, the mechanisms of Alda-1 in hyperoxia-induced ALI remains unclear. C57BL/6 mice implanted with Alzet pumps received Alda-1 in a sustained fashion while being exposed to hyperoxia for 48 h. The mice displayed suppressed immune cell infiltration, decreased protein leakage and alveolar permeability compared to controls. Mechanistic analysis shows that mice pretreated with Alda-1 also experience decreased oxidative stress and enhanced levels of p-Akt and mTOR pathway associated proteins. These results show that continuous delivery of Alda-1 protects against hyperoxia-induced lung injury in mice.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sudarshan Krishnamurthy
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Muling Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Brown School, Washington University, St. Louis, MO, United States
| | | | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
8
|
Feng Y, Wan P, Yin L, Lou X. The Inhibition of MicroRNA-139-5p Promoted Osteoporosis of Bone Marrow-Derived Mesenchymal Stem Cells by Targeting Wnt/Beta-Catenin Signaling Pathway by NOTCH1. J Microbiol Biotechnol 2020; 30:448-458. [PMID: 31752063 PMCID: PMC9728286 DOI: 10.4014/jmb.1908.08036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We investigated the therapeutic effects of microRNA-139-5p in relation to osteoporosis of bone marrow-derived mesenchymal stem cell (BMSCs) and its underlying mechanisms. In this study we used a dexamethasone-induced in vivo model of osteoporosis and BMSCs were used for the in vitro model. Real-time quantitative polymerase chain reaction (RT-PCR) and gene chip were used to analyze the expression of microRNA-139-5p. In an osteoporosis rat model, the expression of microRNA-139-5p was increased, compared with normal group. Downregulation of microRNA-139-5p promotes cell proliferation and osteogenic differentiation in BMSCs. Especially, up-regulation of microRNA-139-5p reduced cell proliferation and osteogenic differentiation in BMSCs. Overexpression of miR-139-5p induced Wnt/β-catenin and down-regulated NOTCH1 signaling in BMSCs. Down-regulation of miR-139-5p suppressed Wnt/β-catenin and induced NOTCH1 signaling in BMSCs. The inhibition of NOTCH1 reduced the effects of anti-miR-139-5p on cell proliferation and osteogenic differentiation in BMSCs. Activation of Wnt/β-catenin also inhibited the effects of anti-miR-139-5p on cell proliferation and osteogenic differentiation in BMSCs. Taken together, our results suggested that the inhibition of microRNA-139-5p promotes osteogenic differentiation of BMSCs via targeting Wnt/β-catenin signaling pathway by NOTCH1.
Collapse
Affiliation(s)
- Yimiao Feng
- Department of Orthodontics, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 30009, P.R. China
| | - Pengbo Wan
- Shangqiu Medical College, Shangqiu, Henan Province 476100, P.R. China
| | - Linling Yin
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (originally named “Shanghai First People’s Hospital”) Shanghai 200080, P.R. China,Corresponding author Phone: +86-21-63240090 E-mail:
| | - Xintian Lou
- Department of Dentistry, Punan Hospital of Pudong New District, Shanghai 200125, P.R. China
| |
Collapse
|
9
|
Liu D, Yuan J, Fei X, Zhu Y, Zhou Y, Zhang C, Dong L, Zhu Z. Effects of inhalation of sevoflurane at different concentrations on TRPV1 in airways of rats at different developmental stages. Life Sci 2020; 249:117472. [PMID: 32112870 DOI: 10.1016/j.lfs.2020.117472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 10/24/2022]
Abstract
Aim Determine changes in the expressions of the ion channel-TRPV1-and neuropeptides-NKA, NKB, calcitonin gene-related peptide (CGRP), and SP-in 14-, 21-, and 42-day-old rats after inhaling 1.5% and 2.6% sevoflurane. MAIN METHODS A small in-house inhalation anesthesia chamber was designed to allow 14-, 21-, and 42-day-old rats inhale 1.5% and 2.6% sevoflurane, and rats in the control group inhaled carrier gas(1 L/min air +1 L/min O2). In addition, 14- and 21-day-old rats were pretreated with capsazepine, followed by inhalation of 1.5% and 2.6% sevoflurane or the carrier gas. The expression of TRPV1 in lung tissues was detected by Western blotting, whereas the expressions of NKA, NKB, CGRP, and SP in the trachea were detected by immunohistochemistry. KEY FINDINGS After inhalation of 1.5% sevoflurane, the expression of TRPV1 in the lung tissues of 14- and 21-day-old rats was significantly increased compared with that in the control group, which was antagonized by capsazepine pretreatment. Moreover, inhalation of 1.5% sevoflurane markedly increased the expressions of NKA, NKB, CGRP, and SP in the trachea of 21-day-old rats and of NKB, CGRP, and SP in the trachea of 14-day-old rats. The expressions of these molecules were antagonized by capsazepine pretreatment. Conversely, inhalation of 2.6% sevoflurane decreased the expressions of NKA and NKB in the trachea of 42-day-old rats. SIGNIFICANCE Sevoflurane did not upregulate the expression of TRPV1 in the airways of late-developing rats. This anesthetic may have a two-way effect on airways, resulting in considerable effects in pediatric clinical anesthesia management.
Collapse
Affiliation(s)
- Dexing Liu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Yuan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xia Fei
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuhang Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yannan Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chao Zhang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Liang Dong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhaoqiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
10
|
Caputo LS, Campos MIC, Dias HJ, Crotti AEM, Fajardo JB, Vanelli CP, Presto ÁCD, Alves MS, Aarestrup FM, Paula ACC, Da Silva Filho AA, Aarestrup BJV, Pereira OS, Corrêa JODA. Copaiba oil suppresses inflammation in asthmatic lungs of BALB/c mice induced with ovalbumin. Int Immunopharmacol 2020; 80:106177. [PMID: 32007706 DOI: 10.1016/j.intimp.2019.106177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/05/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022]
Abstract
Asthma is a chronic inflammatory disease that represents high hospitalizations and deaths in world. Copaiba oil (CO) is popularly used for relieving asthma symptoms and has already been shown to be effective in many inflammation models. This study aimed to investigate the immunomodulatory relationship of CO in ovalbumin (OVA)-induced allergic asthma. The composition of CO sample analyzed by GC and GC-MS and the toxicity test was performed in mice at doses of 50 or 100 mg/kg (by gavage). After, the experimental model of allergic asthma was induced with OVA and mice were orally treated with CO in two pre-established doses. The inflammatory infiltrate was evaluated in bronchoalveolar lavage fluid (BALF), while cytokines (IL-4, IL-5, IL-17, IFN-γ, TNF-α), IgE antibody and nitric oxide (NO) production was evaluated in BALF and lung homogenate (LH) of mice, together with the histology and histomorphometry of the lung tissue. CO significantly attenuated the number of inflammatory cells in BALF, suppressing NO production and reducing the response mediated by TH2 and TH17 (T helper) cells in both BALF and LH. Histopathological and histomorphometric analysis confirmed that CO significantly reduced the numbers of inflammatory infiltrate in the lung tissue, including in the parenchyma area. Our results indicate that CO has an effective in vivo antiasthmatic effect.
Collapse
Affiliation(s)
- Ludmila S Caputo
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Maria Inês C Campos
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Herbert J Dias
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Av. Bandeirantesn° 3900, 14040-901 Ribeirão Preto, SP, Brazil
| | - Antônio E M Crotti
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Av. Bandeirantesn° 3900, 14040-901 Ribeirão Preto, SP, Brazil
| | - Júlia B Fajardo
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Chislene P Vanelli
- Health Department, Faculty of Medical Sciences and Health of Juiz de Fora (SUPREMA), Alameda Salvaterra n° 200, Salvaterra, 36.033-003 Juiz de Fora, MG, Brazil
| | - Álvaro C D Presto
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Maria S Alves
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Fernando M Aarestrup
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Ana Claudia C Paula
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Ademar A Da Silva Filho
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Beatriz J V Aarestrup
- Laboratory of Experimental Immunology and Pathology, Reproduction Biology Center (CBR), Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - Olavo S Pereira
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil
| | - José Otávio do A Corrêa
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Federal University of Juiz de Fora, R. José Lourenço Kelmer s/n, Campus Universitário, 36036-900 Juiz de Fora, MG, Brazil.
| |
Collapse
|
11
|
Chen CM, Hwang J, Chou HC. Maternal Tn Immunization Attenuates Hyperoxia-Induced Lung Injury in Neonatal Rats Through Suppression of Oxidative Stress and Inflammation. Front Immunol 2019; 10:681. [PMID: 31019509 PMCID: PMC6458300 DOI: 10.3389/fimmu.2019.00681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/12/2019] [Indexed: 11/13/2022] Open
Abstract
Hyperoxia therapy is often required to treat newborns with respiratory disorders. Prolonged hyperoxia exposure increases oxidative stress and arrests alveolar development in newborn rats. Tn antigen is N-acetylgalactosamine residue that is one of the most remarkable tumor-associated carbohydrate antigens. Tn immunization increases the serum anti-Tn antibody titers and attenuates hyperoxia-induced lung injury in adult mice. We hypothesized that maternal Tn immunizations would attenuate hyperoxia-induced lung injury through the suppression of oxidative stress in neonatal rats. Female Sprague-Dawley rats (6 weeks old) were intraperitoneally immunized five times with Tn (50 μg/dose) or carrier protein at biweekly intervals on 8, 6, 4, 2, and 0 weeks before the day of delivery. The pups were reared in room air (RA) or 2 weeks of 85% O2, creating the four study groups: carrier protein + RA, Tn vaccine + RA, carrier protein + O2, and Tn vaccine + O2. The lungs were excised for oxidative stress, cytokine, vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) expression, and histological analysis on postnatal day 14. Blood was withdrawn from dams and rat pups to check anti-Tn antibody using western blot. We observed that neonatal hyperoxia exposure reduced the body weight, increased 8-hydroxy-2-deoxyguanosine (8-OHdG) expression and lung cytokine (interleukin-4), increased mean linear intercept (MLI) values, and decreased vascular density and VEGF and PDGF-B expressions. By contrast, Tn immunization increased maternal and neonatal serum anti-Tn antibody titers on postnatal day 14, reduced MLI, and increased vascular density and VEGF and PDGF-B expressions to normoxic levels. Furthermore, the alleviation of lung injury was accompanied by a reduction in lung cytokine and 8-OHdG expression. Therefore, we propose that maternal Tn immunization attenuates hyperoxia-induced lung injury in neonatal rats through the suppression of oxidative stress and inflammation.
Collapse
Affiliation(s)
- Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jaulang Hwang
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Bezerra FS, Ramos CDO, Castro TDF, Araújo NPDS, de Souza ABF, Bandeira ACB, Costa GDP, Cartelle CT, Talvani A, Cangussú SD, Brochard L, Nagato AC. Exogenous surfactant prevents hyperoxia-induced lung injury in adult mice. Intensive Care Med Exp 2019; 7:19. [PMID: 30919149 PMCID: PMC6437243 DOI: 10.1186/s40635-019-0233-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 01/04/2023] Open
Abstract
Background In addition to the risk of developing ventilator-induced lung injury, patients with ARDS are at risk of developing hyperoxic injury due the supra-physiological oxygen supplementation clinically required to reverse hypoxemia. Alterations of endogenous surfactant system participate in the pulmonary dysfunction observed in ARDS. Administration of exogenous surfactant could have protective effects during hyperoxia. Methods Male BALB/c mice (8–10 weeks), a strain highly sensitive to hyperoxia, received the exogenous surfactant-containing protein SP-B and SP-C by intranasal instillation 12 h before starting 24 h of exposure to hyperoxia in an inhalation chamber and were compared to mice receiving hyperoxia alone and to controls subjected to normoxia. Results Compared to the hyperoxia group, the administration of exogenous surfactant was able to reduce lung inflammation through a reduction in the influx of neutrophils and inflammatory biomarkers such as TNF, IL-17, and HMGB1 expression. The antioxidant activity prevented oxidative damage by reducing lipid peroxidation and protein carbonylation and increasing superoxide dismutase activity when compared to the hyperoxia group. Conclusion Our results offer new perspectives on the effects and the mechanism of exogenous surfactant in protecting the airway and lungs, in oxygen-rich lung microenvironment, against oxidative damage and aggravation of acute inflammation induced by hyperoxia.
Collapse
Affiliation(s)
- Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil. .,Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada. .,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada. .,Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of exact and biological sciences (ICEB), Federal University of Ouro Preto (UFOP), Campus Universitário s/n, Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil.
| | - Camila de Oliveira Ramos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Natália Pereira da Silva Araújo
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Ana Carla Balthar Bandeira
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Christiane Teixeira Cartelle
- Laboratory of Neuro Immuno experimental pathology (NIPE), Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Brazil
| | - Laurent Brochard
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Akinori Cardozo Nagato
- Laboratory of Immunopathology and Experimental Pathology, Center for Reproductive Biology-CRB, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.,Physiology Department, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
13
|
Ma X, Hu B, Zou C, Han A, Xu Z, Zhang T, Yu W. The effects of hyperoxia liquid regulate cardiopulmonary bypass‑induced myocardial damage through the Nrf2‑ARE signaling pathway. Mol Med Rep 2018; 18:2342-2348. [PMID: 29901143 DOI: 10.3892/mmr.2018.9162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/06/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the protective role of hyperoxia liquid in regulating cardiopulmonary bypass (CPB)‑induced myocardial damage and its possible underlying mechanism. In the CPB‑induced rat model, hyperoxia liquid enhanced left ventricular ejection fraction (LVEF), reduced the left ventricular internal dimension systole (LVIDs) level, inhibited malondialdehyde levels, increased superoxide dismutase, glutathione (GSH) and GSH peroxidase levels, suppressed heart cell apoptosis, and induced the nuclear factor erythroid 2‑related factor 2 (Nrf2) and heme oxygenase‑1 (HO‑1) signaling pathway. Then, ML385, a Nrf2 inhibitor, was used to attenuate the effect of hyperoxia liquid on LVEF and LVIDs levels, oxidative stress and heart cell apoptosis in the CPB‑induced rat model. Collectively, the results of the present study demonstrated that the protective role of hyperoxia liquid may regulate oxidative stress in a CPB‑induced rat model through the Nrf2‑antioxidant response element signaling pathway.
Collapse
Affiliation(s)
- Xiaochun Ma
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Bin Hu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Aiguo Han
- Department of Clinical Laboratory, The Fifth People's Hospital of Jinan, Jinan, Shandong 250021, P.R. China
| | - Zhenqiang Xu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Tao Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Wancheng Yu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
14
|
Fontes LB, Dias DDS, Aarestrup BJ, Aarestrup FM, Da Silva Filho AA, Corrêa JODA. β -Caryophyllene ameliorates the development of experimental autoimmune encephalomyelitis in C57BL/6 mice. Biomed Pharmacother 2017; 91:257-264. [DOI: 10.1016/j.biopha.2017.04.092] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/25/2023] Open
|
15
|
Yi WJ, Kim TS. Melatonin protects mice against stress-induced inflammation through enhancement of M2 macrophage polarization. Int Immunopharmacol 2017; 48:146-158. [DOI: 10.1016/j.intimp.2017.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/18/2017] [Accepted: 05/06/2017] [Indexed: 01/08/2023]
|
16
|
Helmerhorst HJF, Schouten LRA, Wagenaar GTM, Juffermans NP, Roelofs JJTH, Schultz MJ, de Jonge E, van Westerloo DJ. Hyperoxia provokes a time- and dose-dependent inflammatory response in mechanically ventilated mice, irrespective of tidal volumes. Intensive Care Med Exp 2017; 5:27. [PMID: 28550659 PMCID: PMC5446430 DOI: 10.1186/s40635-017-0142-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/18/2017] [Indexed: 01/26/2023] Open
Abstract
Background Mechanical ventilation and hyperoxia have the potential to independently promote lung injury and inflammation. Our purpose was to study both time- and dose-dependent effects of supplemental oxygen in an experimental model of mechanically ventilated mice. Methods Healthy male C57Bl/6J mice, aged 9–10 weeks, were intraperitoneally anesthetized and randomly assigned to the mechanically ventilated group or the control group. In total, 100 mice were tracheotomized and mechanically ventilated for either 8 or 12 h after allocation to different settings for the applied fractions of inspired oxygen (FiO2, 30, 50, or 90%) and tidal volumes (7.5 or 15 ml/kg). After euthanisation arterial blood, bronchoalveolar lavage fluid (BALf) and tissues were collected for analyses. Results Mechanical ventilation significantly increased the lung injury score (P < 0.05), mean protein content (P < 0.001), and the mean number of cells (P < 0.01), including neutrophils in BALf (P < 0.001). In mice ventilated for 12 h, a significant increase in TNF-α, IFN-γ, IL-1β, IL-10, and MCP-1 (P < 0.01) was observed with 90% FiO2, whereas IL-6 showed a decreasing trend (P for trend = 0.03) across FiO2 groups. KC, MIP-2, and sRAGE were similar between FiO2 groups. HMGB-1 was significantly higher in BALf of mechanically ventilated mice compared to controls and showed a gradual increase in expression with increasing FiO2. Cytokine and chemokine levels in BALf did not markedly differ between FiO2 groups after 8 h of ventilation. Differences between the tidal volume groups were small and did not appear to significantly interact with the oxygen levels. Conclusions We demonstrated a severe vascular leakage and a pro-inflammatory pulmonary response in mechanically ventilated mice, which was enhanced by severe hyperoxia and longer duration of mechanical ventilation. Prolonged ventilation with high oxygen concentrations induced a time-dependent immune response characterized by elevated levels of neutrophils, cytokines, and chemokines in the pulmonary compartment. Electronic supplementary material The online version of this article (doi:10.1186/s40635-017-0142-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hendrik J F Helmerhorst
- Department of Intensive Care Medicine, Leiden University Medical Center, Post Box 9600, 2300 RC, Leiden, The Netherlands. .,Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands. .,Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Amsterdam, The Netherlands.
| | - Laura R A Schouten
- Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Intensive Care Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Gerry T M Wagenaar
- Department of Pediatrics, Laboratory of Neonatology, University Medical Center Leiden, Leiden, The Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Intensive Care Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Marcus J Schultz
- Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Amsterdam, The Netherlands.,Department of Intensive Care Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Evert de Jonge
- Department of Intensive Care Medicine, Leiden University Medical Center, Post Box 9600, 2300 RC, Leiden, The Netherlands
| | - David J van Westerloo
- Department of Intensive Care Medicine, Leiden University Medical Center, Post Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
17
|
The Effects of the Combination of a Refined Carbohydrate Diet and Exposure to Hyperoxia in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1014928. [PMID: 28018521 PMCID: PMC5153507 DOI: 10.1155/2016/1014928] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 12/28/2022]
Abstract
Obesity is a multifactorial disease with genetic, social, and environmental influences. This study aims at analyzing the effects of the combination of a refined carbohydrate diet and exposure to hyperoxia on the pulmonary oxidative and inflammatory response in mice. Twenty-four mice were divided into four groups: control group (CG), hyperoxia group (HG), refined carbohydrate diet group (RCDG), and refined carbohydrate diet + hyperoxia group (RCDHG). The experimental diet was composed of 10% sugar, 45% standard diet, and 45% sweet condensed milk. For 24 hours, the HG and RCDHG were exposed to hyperoxia and the CG and RCDG to ambient air. After the exposures were completed, the animals were euthanized, and blood, bronchoalveolar lavage fluid, and lungs were collected for analyses. The HG showed higher levels of interferon-γ in adipose tissue as compared to other groups and higher levels of interleukin-10 and tumor necrosis factor-α compared to the CG and RCDHG. SOD and CAT activities in the pulmonary parenchyma decreased in the RCDHG as compared to the CG. There was an increase of lipid peroxidation in the HG, RCDG, and RCDHG as compared to the CG. A refined carbohydrate diet combined with hyperoxia promoted inflammation and redox imbalance in adult mice.
Collapse
|
18
|
Shivanna B, Maity S, Zhang S, Patel A, Jiang W, Wang L, Welty SE, Belmont J, Coarfa C, Moorthy B. Gene Expression Profiling Identifies Cell Proliferation and Inflammation as the Predominant Pathways Regulated by Aryl Hydrocarbon Receptor in Primary Human Fetal Lung Cells Exposed to Hyperoxia. Toxicol Sci 2016; 152:155-68. [PMID: 27103661 DOI: 10.1093/toxsci/kfw071] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Exposure to hyperoxia contributes to the development of bronchopulmonary dysplasia (BPD) in premature infants. We observed that aryl hydrocarbon receptor (AhR) signaling protects newborn mice and primary fetal human pulmonary microvascular endothelial cells (HPMECs) against hyperoxic injury. Additionally, a recent genome-wide transcriptome study in a newborn mouse model of BPD identified AhR as a key regulator of hyperoxia-induced gene dysregulation. Whether the AhR similarly deregulates genes in HPMEC is unknown. Therefore, the objective of this study was to characterize transcriptome level gene expression profile in AhR-sufficient and -deficient HPMEC exposed to normoxic and hyperoxic conditions. Global gene expression profiling was performed using Illumina microarray platform and selected genes were validated by real-time RT-PCR. AhR gene expression and hyperoxia independently affected the expression of 540 and 593 genes, respectively. Two-way ANOVA further identified 85 genes that were affected by an interaction between AhR expression and exposure to hyperoxia. Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology, and Reactome pathway analysis identified cell proliferation, immune function, cytokine signaling, and organ development as the major pathways affected in AhR-deficient cells. The biological processes that were significantly enriched by hyperoxia included metabolic process, stress response, signal transduction, cell cycle, and immune regulation. Cell cycle was the predominant pathway affected by the combined effect of AhR knockdown and hyperoxia. Functional analysis of cell cycle showed that AhR-deficient cells had decreased proliferation compared with AhR-sufficient cells. These findings suggest that AhR modulates hyperoxic lung injury by regulating the genes that are necessary for cell proliferation and inflammation.
Collapse
Affiliation(s)
- Binoy Shivanna
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine; *Department of Pediatrics, Section of Neonatal-Perinatal Medicine;
| | | | - Shaojie Zhang
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine
| | - Ananddeep Patel
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine
| | - Weiwu Jiang
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine
| | - Lihua Wang
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine
| | - Stephen E Welty
- *Department of Pediatrics, Section of Neonatal-Perinatal Medicine
| | - John Belmont
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | | | | |
Collapse
|