1
|
Young K, Benny M, Schmidt A, Wu S. Unveiling the Emerging Role of Extracellular Vesicle-Inflammasomes in Hyperoxia-Induced Neonatal Lung and Brain Injury. Cells 2024; 13:2094. [PMID: 39768185 PMCID: PMC11674922 DOI: 10.3390/cells13242094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Extremely premature infants are at significant risk for developing bronchopulmonary dysplasia (BPD) and neurodevelopmental impairment (NDI). Although BPD is a predictor of poor neurodevelopmental outcomes, it is currently unknown how BPD contributes to brain injury and long-term NDI in pre-term infants. Extracellular vesicles (EVs) are small, membrane-bound structures released from cells into the surrounding environment. EVs are involved in inter-organ communication in diverse pathological processes. Inflammasomes are large, multiprotein complexes that are part of the innate immune system and are responsible for triggering inflammatory responses and cell death. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is pivotal in inflammasome assembly and activating inflammatory caspase-1. Activated caspase-1 cleaves gasdermin D (GSDMD) to release a 30 kD N-terminal domain that can form membrane pores, leading to lytic cell death, also known as pyroptosis. Activated caspase-1 can also cleave pro-IL-1β and pro-IL-18 to their active forms, which can be rapidly released through the GSDMD pores to induce inflammation. Recent evidence has emerged that activation of inflammasomes is associated with neonatal lung and brain injury, and inhibition of inflammasomes reduces hyperoxia-induced neonatal lung and brain injury. Additionally, multiple studies have demonstrated that hyperoxia stimulates the release of lung-derived EVs that contain inflammasome cargos. Adoptive transfer of these EVs into the circulation of normal neonatal mice and rats induces brain inflammatory injury. This review focuses on EV-inflammasomes' roles in mediating lung-to-brain crosstalk via EV-dependent and EV-independent mechanisms critical in BPD, brain injury, and NDI pathogenesis. EV-inflammasomes will be discussed as potential therapeutic targets for neonatal lung and brain injury.
Collapse
Affiliation(s)
| | | | | | - Shu Wu
- Division of Neonatology, Department of Pediatrics, Batchelor Children Research Institute, University of Miami School of Medicine, Miami, FL 33136, USA; (K.Y.); (M.B.); (A.S.)
| |
Collapse
|
2
|
Yang X, Wang X, Dong W. Aryl hydrocarbon receptor (AhR) is regulated by hyperoxia in premature infants. J Matern Fetal Neonatal Med 2024; 37:2349179. [PMID: 38816997 DOI: 10.1080/14767058.2024.2349179] [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: 01/22/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE To investigate whether aryl hydrocarbon receptor (AhR) is involved in hyperoxia-mediated oxidative stress by observing the relationship between AhR and reactive oxygen species (ROS) in peripheral blood mononuclear cells (PBMCs) after oxygen exposure in premature infants. METHODS After 48 h of oxygen inhalation at different concentrations, discarded peripheral blood was collected to separate PBMCs and plasma. ROS were labeled with MitoSOXTM Red and detected by fluorescence microscopy in PBMCs. The level of MDA in plasma was detected by thiobarbituric acid colorimetry, the level of MCP-1 in plasma was detected by enzyme-linked immunosorbent assay (ELISA), the localization of AhR was detected by immunofluorescence, and the level of AhR expression in PBMCs was detected by Western blotting. RESULTS As the volume fraction of inspired oxygen increased, compared with those in the air control group, the levels of ROS, MDA in plasma, and MCP-1 in plasma increased gradually in the low concentration oxygen group, medium concentration oxygen group and high concentration oxygen group. The cytoplasm-nuclear translocation rate of AhR gradually increased, and the expression level of AhR gradually decreased. The levels of ROS in PBMCs, MDA in the plasma and MCP-1 in the plasma of premature infants were positively correlated with the cytoplasm-nuclear translocation rate of AhR but negatively correlated with the level of AhR expression. CONCLUSION Aryl hydrocarbon receptor (AhR) is regulated by hyperoxia in premature infants.
Collapse
Affiliation(s)
- Xi Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Xia Wang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| |
Collapse
|
3
|
Tran TT, Davies J, Johnston RA, Karmouty-Quintana H, Li H, Crocker CE, Khan AM, Alcorn JL. Impact of vitamin D on hyperoxic acute lung injury in neonatal mice. BMC Pulm Med 2024; 24:584. [PMID: 39587520 PMCID: PMC11587781 DOI: 10.1186/s12890-024-03391-1] [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: 08/08/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Prolonged exposure to hyperoxia can lead to hyperoxic acute lung injury (HALI) in preterm neonates. Vitamin D (VitD) stimulates lung maturation and acts as an anti-inflammatory agent. Our objective was to determine if VitD provides a dose-dependent protective effect against HALI by reducing inflammatory cytokine expression and improving alveolarization and lung function in neonatal mice. METHODS C57BL/6 mouse neonates were randomized and placed in room air or hyperoxic (85% O2) conditions for 6 days. Control, low (5 ng/neonate) and high (25 ng/neonate) doses of VitD were administered daily beginning at day 2 via oral gavage. Lung tissue was analyzed for edema, changes in pulmonary structure and function, and inflammatory cytokine expression. RESULTS Neonatal mice treated with VitD in hyperoxic conditions had improved weight gain, reduced pulmonary edema and increased alveolar surface area compared to untreated pups in hyperoxia. No significant changes in cytokine expression were observed between untreated and VitD neonates. While changes in surfactant protein mRNA expression were impacted by hyperoxia and VitD administration, no significant changes in alveolar type II cell percentages were observed. At 3 weeks, mice in hyperoxia treated with VitD had greater lung compliance, diminished airway reactivity and improved weight gain. CONCLUSIONS High dose VitD significantly limited harmful effects of HALI. These results suggest that supplementation of VitD to neonatal mice during hyperoxia exposure provides both short-term and long-term protective benefits against HALI.
Collapse
Affiliation(s)
- Thu T Tran
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Jonathan Davies
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard A Johnston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 3.230B, Houston, TX, 77030, USA
| | - Huiling Li
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Caroline E Crocker
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Amir M Khan
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Joseph L Alcorn
- Division of Neonatology, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
4
|
Ji C, Hao X, Li Z, Liu J, Yan H, Ma K, Li L, Zhang L. Phillyrin prevents sepsis-induced acute lung injury through inhibiting the NLRP3/caspase-1/GSDMD-dependent pyroptosis signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39394820 DOI: 10.3724/abbs.2024161] [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: 10/14/2024] Open
Abstract
Acute lung injury (ALI) is a severe pulmonary disorder of sepsis with high clinical incidence and mortality. Nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3)-cysteinyl aspartate specific proteinase 1-gasdermin D (GSDMD)-dependent pyroptosis of alveolar epithelial cells (AECs) has emerged as a crucial contributor to ALI during sepsis. Phillyrin (PHI), a natural lignan isolated from the traditional Chinese herbal medicine Forsythia suspensa, has been shown to have anti-inflammatory, antioxidant and antiviral properties. However, little is known about the protective role and potential mechanism of PHI in sepsis-induced ALI, and it is uncertain whether the protective effect of PHI in sepsis-induced ALI is connected to pyroptosis. This study aims to examine the preventive effects of PHI on sepsis-induced ALI via the inhibition of NLRP3/caspase-1/GSDMD-mediated pyroptosis in AECs. Our findings demonstrate that preadministration of PHI successfully reduces sepsis-induced pulmonary edema, systemic/pulmonary inflammation, and pulmonary histological damage in lung tissues, bronchoalveolar lavage fluid, and the serum of septic mice. Intriguingly, PHI preadministration suppresses sepsis-induced protein expressions of pyroptosis-specific markers, especially their active forms. In vitro assays show that PHI pretreatment also protects type II AECs (MLE-12) from lipopolysaccharide-induced pyroptosis by preventing the activation of the pyroptosis signaling pathway. The results from molecular docking and surface plasmon resonance reveal that PHI has a significant affinity for direct binding to the GSDMD protein, suggesting that GSDMD is a potential pharmacological target for PHI. In conclusion, PHI can prevent sepsis-triggered ALI by effectively suppressing the activation of the canonical pyroptosis signaling pathway and pyroptosis of AECs.
Collapse
Affiliation(s)
- Chen Ji
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
| | - Xiaoyan Hao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
| | - Zhiyi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
| | - Jiaxing Liu
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
| | - Hanyu Yan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, the first Affiliated Hospital, Shihezi University, Shihezi 832008, China
| | - Ling Li
- Medical Teaching Experimental Center, School of Medicine, Shihezi University, Shihezi 832003, China
| | - Liang Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, the first Affiliated Hospital, Shihezi University, Shihezi 832008, China
| |
Collapse
|
5
|
Chen D, Man LY, Wang YY, Zhu WY, Zhao HM, Li SP, Zhang YL, Li SC, Wu YX, Ling-Ai, Pang QF. Nrf2 deficiency exacerbated pulmonary pyroptosis in maternal hypoxia-induced intrauterine growth restriction offspring mice. Reprod Toxicol 2024; 129:108671. [PMID: 39038764 DOI: 10.1016/j.reprotox.2024.108671] [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: 05/20/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
Maternal prenatal hypoxia is an important contributor to intrauterine growth restriction (IUGR), which impedes fetal lung maturation and leads to the development of chronic lung diseases. Although evidence suggests the involvement of pyroptosis in IUGR, the molecular mechanism of pyroptosis is still unclear. Nuclear factor erythroid 2-related factor 2 (Nrf2) has been found to potentially interact with gasdermin D (GSDMD), the key protein responsible for pyroptosis, indicating its crucial role in inhibiting pyroptosis. Therefore, we hypothesized that Nrf2 deficiency is a key molecular responsible for lung pyroptosis in maternal hypoxia-induced IUGR offspring mice. Pregnant WT and Nrf2-/- mice were exposed to hypoxia (10.5 % O2) to mimic IUGR model. We assessed body weight, lung histopathology, pulmonary angiogenesis, oxidative stress levels, as well as mRNA and protein expressions related to inflammation in the 2-week-old offspring. Additionally, we conducted a dual-luciferase reporter assay to confirm the targeting relationship between Nrf2 and GSDMD. Our findings revealed that offspring with maternal hypoxia-induced IUGR exhibited reduced birth weight, catch-up growth delay, and pulmonary dysplasia. Furthermore, we observed impaired nuclear translocation of Nrf2 and increased GSDMD-mediated pyroptosis in these offspring with IUGR. Moreover, the dual-luciferase reporter assay demonstrated that Nrf2 could directly inhibit GSDMD transcription; deficiency of Nrf2 exacerbated pyroptosis and pulmonary dysplasia in offspring with maternal hypoxia-induced IUGR. Collectively, our findings suggest that Nrf2 deficiency induces GSDMD-mediated pyroptosis and pulmonary dysplasia in offspring with maternal hypoxia-induced IUGR; thus highlighting the potential therapeutic approach of targeting Nrf2 for treating prenatal hypoxia-induced pulmonary dysplasia in offspring.
Collapse
Affiliation(s)
- Dan Chen
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Ling-Yun Man
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Ying-Ying Wang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Wei-Ying Zhu
- Department of obstetric, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China
| | - Hui-Min Zhao
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Sheng-Peng Li
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Yan-Li Zhang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Shuai-Chao Li
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Ya-Xian Wu
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China
| | - Ling-Ai
- Department of obstetric, Maternity and Child Health Care Affiliated Hospital, Jiaxing University, Jiaxing 314000, China.
| | - Qing-Feng Pang
- Department of physiopathology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu Province 214122, China.
| |
Collapse
|
6
|
Rao T, Zhou Y, Chen C, Chen J, Zhang J, Lin W, Jia D. Recent progress in neonatal hyperoxic lung injury. Pediatr Pulmonol 2024; 59:2414-2427. [PMID: 38742254 DOI: 10.1002/ppul.27062] [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: 12/13/2023] [Revised: 03/28/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
With the progress in neonatal intensive care, there has been an increase in the survival rates of premature infants. However, this has also led to an increased incidence of neonatal hyperoxia lung injury and bronchopulmonary dysplasia (BPD), whose pathogenesis is believed to be influenced by various prenatal and postnatal factors, although the exact mechanisms remain unclear. Recent studies suggest that multiple mechanisms might be involved in neonatal hyperoxic lung injury and BPD, with sex also possibly playing an important role, and numerous drugs have been proposed and shown promise for improving the treatment outcomes of hyperoxic lung injury. Therefore, this paper aims to analyze and summarize sex differences in neonatal hyperoxic lung injury, potential pathogenesis and treatment progress to provide new ideas for basic and clinical research in this field.
Collapse
Affiliation(s)
- Tian Rao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiyang Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chizhang Chen
- Department of Clinical Medicine, Chinese Medicine Hospital of Pingyang, Wenzhou, Zhejiang, China
| | - Jiayi Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Endesfelder S. Caffeine: The Story beyond Oxygen-Induced Lung and Brain Injury in Neonatal Animal Models-A Narrative Review. Antioxidants (Basel) 2024; 13:1076. [PMID: 39334735 PMCID: PMC11429035 DOI: 10.3390/antiox13091076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Caffeine is one of the most commonly used drugs in intensive care to stimulate the respiratory control mechanisms of very preterm infants. Respiratory instability, due to the degree of immaturity at birth, results in apnea of prematurity (AOP), hyperoxic, hypoxic, and intermittent hypoxic episodes. Oxidative stress cannot be avoided as a direct reaction and leads to neurological developmental deficits and even a higher prevalence of respiratory diseases in the further development of premature infants. Due to the proven antioxidant effect of caffeine in early use, largely protective effects on clinical outcomes can be observed. This is also impressively observed in experimental studies of caffeine application in oxidative stress-adapted rodent models of damage to the developing brain and lungs. However, caffeine shows undesirable effects outside these oxygen toxicity injury models. This review shows the effects of caffeine in hyperoxic, hypoxic/hypoxic-ischemic, and intermittent hypoxic rodent injury models, but also the negative effects on the rodent organism when caffeine is administered without exogenous oxidative stress. The narrative analysis of caffeine benefits in cerebral and pulmonary preterm infant models supports protective caffeine use but should be given critical consideration when considering caffeine treatment beyond the recommended corrected gestational age.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
8
|
Zou D, Liao J, Xiao M, Liu L, Xu M. Melatonin alleviates hyperoxia-induced lung injury through elevating MSC exosomal miR-18a-5p expression to repress PUM2 signaling. FASEB J 2024; 38:e70012. [PMID: 39183539 DOI: 10.1096/fj.202400374r] [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: 02/18/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Mesenchymal stem cells (MSC)-derived exosomes (Exo) are a possible option for hyperoxia-induced lung injury (HLI). We wanted to see if melatonin (MT)-pretreated MSC-derived exosomes (MT-Exo) were more effective against HLI, and we also tried to figure out the underlying mechanism. HLI models were established by hyperoxia exposure. HE staining was adopted to analyze lung pathological changes. MTT and flow cytometry were used to determine cell viability and apoptosis, respectively. The mitochondrial membrane potential (MMP) was analyzed using the JC-1 probe. LDH, ROS, SOD, and GSH-Px levels were examined by the corresponding kits. The interactions between miR-18a-5p, PUM2, and DUB3 were analyzed by molecular interaction experiments. MT-Exo could effectively inhibit hyperoxia-induced oxidative stress, inflammatory injury, and apoptosis in lung epithelial cells, while these effects of MT-Exo were weakened by miR-18a-5p knockdown in MSCs. miR-18a-5p reduced PUM2 expression in MLE-12 cells by directly targeting PUM2. In addition, PUM2 inactivated the Nrf2/HO-1 signaling pathway by promoting DUB3 mRNA decay post-transcriptionally. As expected, PUM2 overexpression or DUB3 knockdown abolished the protective effect of MT-Exo on hyperoxia-induced lung epithelial cell injury. MT-Exo carrying miR-18a-5p reduced hyperoxia-mediated lung injury in mice through activating Nrf2/HO-1 pathway. MT reduced PUM2 expression and subsequently activated the DUB3/Nrf2/HO-1 signal axis by increasing miR-18a-5p expression in MSC-derived exosomes to alleviate HLI.
Collapse
Affiliation(s)
- Dongmei Zou
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Jinwen Liao
- The Department of Pediatric, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong, China
| | - Min Xiao
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Liang Liu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
| | - Mingguo Xu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, Guangdong, China
- The Department of Pediatric, The Third People's Hospital of Longgang District Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Lemes Dos Santos Sanna P, Bernardes Carvalho L, Cristina Dos Santos Afonso C, de Carvalho K, Aires R, Souza J, Rodrigues Ferreira M, Birbrair A, Martha Bernardi M, Latini A, Foganholi da Silva RA. Adora2A downregulation promotes caffeine neuroprotective effect against LPS-induced neuroinflammation in the hippocampus. Brain Res 2024; 1833:148866. [PMID: 38494098 DOI: 10.1016/j.brainres.2024.148866] [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: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Caffeine has been extensively studied in the context of CNS pathologies as many researchers have shown that consuming it reduces pro-inflammatory biomarkers, potentially delaying the progression of neurodegenerative pathologies. Several lines of evidence suggest that adenosine receptors, especially A1 and A2A receptors, are the main targets of its neuroprotective action. We found that caffeine pretreatment 15 min before LPS administration reduced the expression of Il1b in the hippocampus and striatum. The harmful modulation of caffeine-induced inflammatory response involved the downregulation of the expression of A2A receptors, especially in the hippocampus. Caffeine treatment alone promoted the downregulation of the adenosinergic receptor Adora2A; however, this promotion effect was reversed by LPS. Although administering caffeine increased the expression of the enzymes DNA methyltransferases 1 and 3A and decreased the expression of the demethylase enzyme Tet1, this effect was reversed by LPS in the hippocampus of mice that were administered Caffeine + LPS, relative to the basal condition; no significant differences were observed in the methylation status of the promoter regions of adenosine receptors. Finally, the bioinformatics analysis of the expanded network demonstrated the following results: the Adora2B gene connects the extended networks of the adenosine receptors Adora1 and Adora2A; the Mapk3 and Esr1 genes connect the extended Adora1 network; the Mapk4 and Arrb2 genes connect the extended Adora2A network with the extended network of the proinflammatory cytokine Il1β. These results indicated that the anti-inflammatory effects of acute caffeine administration in the hippocampus may be mediated by a complex network of interdependencies between the Adora2B and Adora2A genes.
Collapse
Affiliation(s)
| | | | | | - Kassia de Carvalho
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Rogério Aires
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Jennyffer Souza
- Laboratory of Bioenergetics and Oxidative Stress - LABOX, Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Marcel Rodrigues Ferreira
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unity, Botucatu Medical School, São Paulo State University, Brazil.
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA
| | - Maria Martha Bernardi
- Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil
| | - Alexandra Latini
- Laboratory of Bioenergetics and Oxidative Stress - LABOX, Department of Biochemistry, Center for Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rodrigo A Foganholi da Silva
- Dentistry, University of Taubaté, Taubaté, São Paulo, São Paulo, Brazil; Center for Epigenetic Study and Genic Regulation - CEEpiRG, Program in Environmental and Experimental Pathology, Paulista University, São Paulo, São Paulo, Brazil.
| |
Collapse
|
10
|
Yang K, Liu J, He T, Dong W. Caffeine and neonatal acute kidney injury. Pediatr Nephrol 2024; 39:1355-1367. [PMID: 37665410 DOI: 10.1007/s00467-023-06122-6] [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: 04/24/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Acute kidney injury is one of the most threatening diseases in neonates, with complex pathogenesis and limited treatment options. Caffeine is a commonly used central nervous system stimulant for treating apnea in preterm infants. There is compelling evidence that caffeine may have potential benefits for preventing neonatal acute kidney injury, but comprehensive reports are lacking in this area. Hence, this review aims to provide a summary of clinical data on the potential benefits of caffeine in improving neonatal acute kidney injury. Additionally, it delves into the molecular mechanisms underlying caffeine's effects on acute kidney injury, with a focus on various aspects such as oxidative stress, adenosine receptors, mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome, autophagy, p53, and gut microbiota. The ultimate goal of this review is to provide information for healthcare professionals regarding the link between caffeine and neonatal acute kidney injury and to identify gaps in our current understanding.
Collapse
Affiliation(s)
- Kun Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Jinjing Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Ting He
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China.
| |
Collapse
|
11
|
Shen X, Yang Z, Wang Q, Chen X, Zhu Q, Liu Z, Patel N, Liu X, Mo X. Lactobacillus plantarum L168 improves hyperoxia-induced pulmonary inflammation and hypoalveolarization in a rat model of bronchopulmonary dysplasia. NPJ Biofilms Microbiomes 2024; 10:32. [PMID: 38553470 PMCID: PMC10980738 DOI: 10.1038/s41522-024-00504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
Alteration of gut microbiota can affect chronic lung diseases, such as asthma and chronic obstructive pulmonary disease, through abnormal immune and inflammatory responses. Previous studies have shown a feasible connection between gut microbiota and bronchopulmonary dysplasia (BPD) in preterm infants. However, whether BPD can be ameliorated by restoring the gut microbiota remains unclear. In preterm infants with BPD, we found variance in the diversity and structure of gut microbiota. Similarly, BPD rats showed gut dysbiosis, characterized by a deficiency of Lactobacillus, which was abundant in normal rats. We therefore explored the effect and potential mechanism of action of a probiotic strain, Lactobacillus plantarum L168, in improving BPD. The BPD rats were treated with L. plantarum L168 by gavage for 2 weeks, and the effect was evaluated by lung histopathology, lung function, and serum inflammatory markers. Subsequently, we observed reduced lung injury and improved lung development in BPD rats exposed to L. plantarum L168. Further evaluation revealed that L. plantarum L168 improved intestinal permeability in BPD rats. Serum metabolomics showed altered inflammation-associated metabolites following L. plantarum L168 intervention, notably a marked increase in anti-inflammatory metabolites. In agreement with the metabolites analysis, RNA-seq analysis of the intestine and lung showed that inflammation and immune-related genes were down-regulated. Based on the information from RNA-seq, we validated that L. plantarum L168 might improve BPD relating to down-regulation of TLR4 /NF-κB /CCL4 pathway. Together, our findings suggest the potential of L. plantarum L168 to provide probiotic-based therapeutic strategies for BPD.
Collapse
Affiliation(s)
- Xian Shen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qihui Zhu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Zhi Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China.
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Zhang X, Cao R, Li C, Zhao H, Zhang R, Che J, Xie J, Tang N, Wang Y, Liu X, Zheng Q. Caffeine Ameliorates Age-Related Hearing Loss by Downregulating the Inflammatory Pathway in Mice. Otol Neurotol 2024; 45:227-237. [PMID: 38320571 PMCID: PMC10922330 DOI: 10.1097/mao.0000000000004098] [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] [Indexed: 02/08/2024]
Abstract
OBJECTIVE Age-related hearing loss (ARHL), also known as presbycusis, is a debilitating sensory impairment that affects the elderly population. There is currently no ideal treatment for ARHL. Long-term caffeine intake was reported to have anti-aging effects in many diseases. This study is to identify whether caffeine could ameliorate ARHL in mice and analyze its mechanism. METHODS Caffeine was administered in drinking water to C57BL/6J mice from the age of 3 months to 12 months. The body weight, food intake and water intake of the mice were monitored during the experiment. The metabolic indicators of serum were detected by ELISA. The function of the hearing system was evaluated by ABR and hematoxylin and eosin staining of the cochlea. Genes' expression were detected by Q-PCR, immunofluorescencee and Western blot. RESULTS The results showed that the ARHL mice exhibited impaired hearing and cochlear tissue compared with the young mice. However, the caffeine-treated ARHL mice showed improved hearing and cochlear tissue morphology. The expression of inflammation-related genes, such as TLR4, Myd88, NF-κB, and IL-1β, was significantly increased in the cochleae of ARHL mice compared with young mice but was down-regulated in the caffeine-treated cochleae. CONCLUSIONS Inflammation is involved in ARHL of mice, and long-term caffeine supplementation could ameliorate ARHL through the down-regulation of the TLR4/NF-κB inflammation pathway. Our findings provide a new idea for preventing ARHL and suggest new drug targets for ARHL treatment.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Ruijuan Cao
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Changye Li
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Hongchun Zhao
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Ruyi Zhang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Juan Che
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Jinwen Xie
- Shandong Binzhou Animal science and veterinary Medicine Academy, Binzhou, China, 256600
| | - Na Tang
- Shandong Binzhou Animal science and veterinary Medicine Academy, Binzhou, China, 256600
| | - Yanfei Wang
- Department of Otolaryngology/Head and Neck Surgery, Institute of Otolaryngology, Binzhou Medical University Hospital, Binzhou, China
| | - Xiuzhen Liu
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Qingyin Zheng
- Department of Otolaryngology-HNS, Case Western Reserve University
| |
Collapse
|
13
|
Yang M, Chen Y, Huang X, Shen F, Meng Y. Lysine demethylase KDM3A alleviates hyperoxia-induced bronchopulmonary dysplasia in mice by promoting ETS1 expression. Exp Cell Res 2024; 435:113945. [PMID: 38286256 DOI: 10.1016/j.yexcr.2024.113945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 01/31/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease among neonates, with increasing morbidity and mortality. This study aims to investigate the effect and mechanism of lysine demethylase 3A (KDM3A) on hyperoxia-induced BPD. Hyperoxia-induced BPD mouse and alveolar epithelial cell models were constructed. The effects of hyperoxia on lung development were evaluated by histological and morphological analysis. The levels of KDM3A, E26 transformation specific-1 (ETS1), H3 lysine 9 dimethylation (H3K9me2), and endoplasmic reticulum (ER) stress-related indexes were quantified by RT-qPCR, Western blot, and IF staining. Cell apoptosis was assessed by flow cytometry and TUNEL staining. Transfection of oe-ETS1, oe-KDM3A, and sh-ETS1 was applied in hyperoxia-induced alveolar epithelial cells to explore the mechanism of the KDM3A/ETS1 axis in hyperoxia-induced apoptosis. KDM3A inhibitor IOX1 was applied to validate the in vivo effect of KDM3A in hyperoxia-induced BPD mice. The results displayed that hyperoxia-induced BPD mice showed reduced body weight, severe destruction of alveolar structure, decreased radial alveolar count (RAC), and increased mean linear intercept (MLI) and mean alveolar diameter (MAD). Further, hyperoxia induction down-regulated ETS1 expression, raised ER stress levels, and increased apoptosis rate in BPD mice and alveolar epithelial cells. However, transfection of oe-ETS1 improved the above changes in hyperoxia-induced alveolar epithelial cells. Moreover, transfection of oe-KDM3A up-regulated ETS1 expression, down-regulated H3K9me2 expression, inhibited ER stress, and reduced apoptosis rate in hyperoxia-induced alveolar epithelial cells. In addition, transfection of sh-ETS1 reversed the inhibitory effect of KDM3A on hyperoxia-induced apoptosis by regulating ER stress. In vivo experiments, KDM3A inhibitor IOX1 intervention further aggravated BPD in newborn mice. In a word, KDM3A alleviated hyperoxia-induced BPD in mice by promoting ETS1 expression.
Collapse
Affiliation(s)
- Min Yang
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China.
| | - Yanping Chen
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China
| | | | - Fang Shen
- Research Institute of Children, Hunan Children's Hospital, Changsha, 410007, China
| | - Yanni Meng
- Respiratory Department, Hunan Children's Hospital, Changsha, 410007, China
| |
Collapse
|
14
|
Weng W, Wang X, Cui Y. Artesunate Alleviates Chronic Hyperoxia-induced Bronchopulmonary Dysplasia by Suppressing NF-κB Pathway in Neonatal Mice. Comb Chem High Throughput Screen 2024; 27:2681-2690. [PMID: 37861045 DOI: 10.2174/0113862073246710231002042239] [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: 02/23/2023] [Revised: 07/29/2023] [Accepted: 08/30/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a chronic lung condition that occurs in premature infants who undergo prolonged mechanical ventilation and oxygen therapy. Existing treatment methods have shown limited efficacy, highlighting the urgent need for new therapeutic strategies. Artesunate (AS) is a compound known for its potential anti-inflammatory properties, and studies have shown its protective effects against acute lung injury. However, its impact on BPD and the underlying mechanisms remain unclear. OBJECTIVE To investigate the effect and underlying mechanism of AS on chronic hyperoxiainduced BPD in neonatal mice. METHOD Full-term C57BL/6J mice were randomly assigned to the Air+lactate Ringer's solution (L/R) group, O2 + L/R group, and O2 + AS group. Analysis was performed using assay methods such as ELISA, RT-qPCR, hematoxylin-eosin staining, and Western blotting. RESULTS Compared with the O2+L/R group, the expression of inflammatory factors in the serum, tissue, and BALF of the O2+AS group was significantly reduced, the lung function of the mice was improved, and the inflammatory infiltrates were significantly alleviated. AS inhibited the mRNA expression of inflammatory factors in mice. We found that the expression of nuclear p65 and cytoplasmic p-IκBα in the NF-κB pathway was inhibited after adding AS. CONCLUSION AS ameliorated chronic hyperoxia-induced BPD in neonatal mice probably by inhibiting the expression of NF-κB pathway and inflammatory factors.
Collapse
Affiliation(s)
- Wenbo Weng
- Yuyao Health Training School, Ningbo, Zhejiang, China
| | - Xiaoying Wang
- Department of Pediatrics, Jiande First People's Hospital, Hangzhou, Zhejiang, China
| | - Yifei Cui
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Deng MG, Liu F, Wang K, Liang Y, Nie JQ, Chai C. Genetic association between coffee/caffeine consumption and the risk of obstructive sleep apnea in the European population: a two-sample Mendelian randomization study. Eur J Nutr 2023; 62:3423-3431. [PMID: 37668652 DOI: 10.1007/s00394-023-03239-0] [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: 11/17/2022] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND The association between coffee/caffeine consumption and obstructive sleep apnea (OSA) risk remains unclear. PURPOSE To determine the relationship between coffee/caffeine consumption and the risk of OSA, using the Mendelian randomization (MR) method in the European population. METHODS Two sets of coffee consumption-associated genetic variants were, respectively, extracted from the recent genome-wide meta-analysis (GWMA) and genome-wide association study (GWAS) of coffee consumption. Taking other caffeine sources into account, genetic variants associated with caffeine consumption from tea and plasma caffeine (reflecting total caffeine intake) were also obtained. The inverse variance weighted (IVW) technique was utilized as the primary analysis, supplemented by the MR-Egger, weighted-median, and MR-Pleiotropy RESidual Sum and Outlier (PRESSO) techniques. Leave-one-out (LOO) analysis was performed to assess whether the overall casual estimates were driven by a single SNP. Additional sensitivity analyses were performed using similar methods, while the genetic variants associated with confounders, e.g., body mass index and hypertension, were excluded. RESULTS The IVW method demonstrated that coffee consumption GWMA (OR: 1.065, 95% CI 0.927-1.224, p = 0.376), coffee consumption GWAS (OR: 1.665, 95% CI 0.932-2.977, p = 0.086), caffeine from tea (OR: 1.198, 95% CI 0.936-1.534, p = 0.151), and blood caffeine levels (OR: 1.054, 95% CI 0.902-1.231, p = 0.508) were unlikely to be associated with the risk of OSA. The other three methods presented similar results, where no significant associations were found. No single genetic variant was driving the overall estimates by the LOO analysis. These findings were also supported by the sensitivity analyses with no confounding genetic variants. CONCLUSION Our study found no association between coffee/caffeine consumption and the risk of OSA.
Collapse
Affiliation(s)
- Ming-Gang Deng
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei, China.
| | - Fang Liu
- School of Public Health, Wuhan University, Wuhan, 430071, Hubei, China
| | - Kai Wang
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, 430033, Hubei, China
| | - Yuehui Liang
- School of Public Health, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jia-Qi Nie
- Xiaogan Center for Disease Control and Prevention, Xiaogan, 432000, Huebi, China
| | - Chen Chai
- Emergency Center, Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| |
Collapse
|
16
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [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: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
17
|
Aslan M, Gokce IK, Turgut H, Tekin S, Cetin Taslidere A, Deveci MF, Kaya H, Tanbek K, Gul CC, Ozdemir R. Molsidomine decreases hyperoxia-induced lung injury in neonatal rats. Pediatr Res 2023; 94:1341-1348. [PMID: 37179436 DOI: 10.1038/s41390-023-02643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND The study's objective is to evaluate if Molsidomine (MOL), an anti-oxidant, anti-inflammatory, and anti-apoptotic drug, is effective in treating hyperoxic lung injury (HLI). METHODS The study consisted of four groups of neonatal rats characterized as the Control, Control+MOL, HLI, HLI + MOL groups. Near the end of the study, the lung tissue of the rats were evaluated with respect to apoptosis, histopathological damage, anti-oxidant and oxidant capacity as well as degree of inflammation. RESULTS Compared to the HLI group, malondialdehyde and total oxidant status levels in lung tissue were notably reduced in the HLI + MOL group. Furthermore, mean superoxide dismutase, glutathione peroxidase, and glutathione activities/levels in lung tissue were significantly higher in the HLI + MOL group as compared to the HLI group. Tumor necrosis factor-α and interleukin-1β elevations associated with hyperoxia were significantly reduced following MOL treatment. Median histopathological damage and mean alveolar macrophage numbers were found to be higher in the HLI and HLI + MOL groups when compared to the Control and Control+MOL groups. Both values were increased in the HLI group when compared to the HLI + MOL group. CONCLUSIONS Our research is the first to demonstrate that bronchopulmonary dysplasia may be prevented through the protective characteristics of MOL, an anti-inflammatory, anti-oxidant, and anti-apoptotic drug. IMPACT Molsidomine prophylaxis significantly decreased the level of oxidative stress markers. Molsidomine administration restored the activities of antioxidant enzymes. Molsidomine prophylaxis significantly reduced the levels of inflammatory cytokines. Molsidomine may provide a new and promising therapy for BPD in the future. Molsidomine prophylaxis decreased lung damage and macrophage infiltration in the tissue.
Collapse
Affiliation(s)
- Mehmet Aslan
- Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Ismail Kursat Gokce
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Turgut
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Suat Tekin
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Asli Cetin Taslidere
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Fatih Deveci
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Huseyin Kaya
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Kevser Tanbek
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Cemile Ceren Gul
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Ramazan Ozdemir
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey.
| |
Collapse
|
18
|
Vincenzi F, Pasquini S, Contri C, Cappello M, Nigro M, Travagli A, Merighi S, Gessi S, Borea PA, Varani K. Pharmacology of Adenosine Receptors: Recent Advancements. Biomolecules 2023; 13:1387. [PMID: 37759787 PMCID: PMC10527030 DOI: 10.3390/biom13091387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Adenosine receptors (ARs) are widely acknowledged pharmacological targets yet are still underutilized in clinical practice. Their ubiquitous distribution in almost all cells and tissues of the body makes them, on the one hand, excellent candidates for numerous diseases, and on the other hand, intrinsically challenging to exploit selectively and in a site-specific manner. This review endeavors to comprehensively depict the substantial advancements witnessed in recent years concerning the development of drugs that modulate ARs. Through preclinical and clinical research, it has become evident that the modulation of ARs holds promise for the treatment of numerous diseases, including central nervous system disorders, cardiovascular and metabolic conditions, inflammatory and autoimmune diseases, and cancer. The latest studies discussed herein shed light on novel mechanisms through which ARs exert control over pathophysiological states. They also introduce new ligands and innovative strategies for receptor activation, presenting compelling evidence of efficacy along with the implicated signaling pathways. Collectively, these emerging insights underscore a promising trajectory toward harnessing the therapeutic potential of these multifaceted targets.
Collapse
Affiliation(s)
- Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Martina Cappello
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| | | | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (C.C.); (M.C.); (M.N.); (A.T.); (S.M.); (S.G.); (K.V.)
| |
Collapse
|
19
|
Saraiva SM, Jacinto TA, Gonçalves AC, Gaspar D, Silva LR. Overview of Caffeine Effects on Human Health and Emerging Delivery Strategies. Pharmaceuticals (Basel) 2023; 16:1067. [PMID: 37630983 PMCID: PMC10459237 DOI: 10.3390/ph16081067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Caffeine is a naturally occurring alkaloid found in various plants. It acts as a stimulant, antioxidant, anti-inflammatory, and even an aid in pain management, and is found in several over-the-counter medications. This naturally derived bioactive compound is the best-known ingredient in coffee and other beverages, such as tea, soft drinks, and energy drinks, and is widely consumed worldwide. Therefore, it is extremely important to research the effects of this substance on the human body. With this in mind, caffeine and its derivatives have been extensively studied to evaluate its ability to prevent diseases and exert anti-aging and neuroprotective effects. This review is intended to provide an overview of caffeine's effects on cancer and cardiovascular, immunological, inflammatory, and neurological diseases, among others. The heavily researched area of caffeine in sports will also be discussed. Finally, recent advances in the development of novel nanocarrier-based formulations, to enhance the bioavailability of caffeine and its beneficial effects will be discussed.
Collapse
Affiliation(s)
- Sofia M. Saraiva
- CPIRN-UDI/IPG, Center of Potential and Innovation of Natural Resources, Research Unit for Inland Development (UDI), Polytechnic Institute of Guarda, 6300-559 Guarda, Portugal; (S.M.S.); (T.A.J.)
| | - Telma A. Jacinto
- CPIRN-UDI/IPG, Center of Potential and Innovation of Natural Resources, Research Unit for Inland Development (UDI), Polytechnic Institute of Guarda, 6300-559 Guarda, Portugal; (S.M.S.); (T.A.J.)
| | - Ana C. Gonçalves
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6201-001 Covilhã, Portugal;
| | - Dário Gaspar
- Department of Sport Sciences, University of Beira Interior, 6201-001 Covilhã, Portugal;
| | - Luís R. Silva
- CPIRN-UDI/IPG, Center of Potential and Innovation of Natural Resources, Research Unit for Inland Development (UDI), Polytechnic Institute of Guarda, 6300-559 Guarda, Portugal; (S.M.S.); (T.A.J.)
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, 6201-001 Covilhã, Portugal;
- Department of Chemical Engineering, University of Coimbra, CIEPQPF, Rua Sílvio Lima, Pólo II—Pinhal de Marrocos, 3030-790 Coimbra, Portugal
| |
Collapse
|
20
|
He F, Wang QF, Li L, Yu C, Liu CZ, Wei WC, Chen LP, Li HY. Melatonin Protects Against Hyperoxia-Induced Apoptosis in Alveolar Epithelial type II Cells by Activating the MT2/PI3K/AKT/ETS1 Signaling Pathway. Lung 2023; 201:225-234. [PMID: 36928143 DOI: 10.1007/s00408-023-00610-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE Hyperoxia-induced apoptosis in alveolar epithelial type II cells (AECIIs) plays a critical role in the development of bronchopulmonary dysplasia (BPD). Melatonin has been shown to improve BPD. However, the protective effect of melatonin on hyperoxia-induced apoptosis in AECIIs and the precise mechanisms involved remain unclear. METHODS Human alveolar epithelial type II A549 cells were treated with hyperoxia as an in vitro model to investigate the antiapoptotic mechanism of melatonin. CCK-8 assays were performed to investigate the viability of A549 cells. Hoechst 33,258 staining was carried out to quantify apoptosis in A549 cells. The protein expression levels of E26 oncogene homolog 1 (ETS1), Bcl-2, Bax, Bim, Wnt, β-catenin, AKT and phosphorylated AKT were measured by western blotting. LY294002, SC79 and the downregulation of ETS1, melatonin receptor 1 (MT1) and MT2 with specific siRNAs were used to investigate the role of the PI3K/AKT pathway, ETS1, MT1 and MT2 in hyperoxia-induced apoptosis in A549 cells. RESULTS Melatonin prevented hyperoxia-induced apoptosis in A549 cells, and the upregulation of E26 oncogene homolog 1 (ETS1) contributed to the antiapoptotic effect of melatonin. Melatonin activated the PI3K/AKT axis, which led to ETS1 upregulation and inhibited apoptosis in hyperoxia-exposed A549 cells. Furthermore, melatonin-induced activation of the PI3K/AKT axis, upregulation of ETS1 and inhibition of apoptosis were reversed by melatonin receptor 2 (MT2) siRNA in hyperoxia-exposed A549 cells. CONCLUSION Melatonin prevents hyperoxia-induced apoptosis by activating the MT2/PI3K/AKT/ETS1 axis in alveolar epithelial cells.
Collapse
Affiliation(s)
- Fan He
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Qiao-Fang Wang
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Lin Li
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Cong Yu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Chun-Zi Liu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Wen-Chao Wei
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Li-Ping Chen
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China.
| | - Huan-Yu Li
- Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No. 152, Aiguo Road, DongHu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
21
|
Artesunate Alleviates Hyperoxia-Induced Lung Injury in Neonatal Mice by Inhibiting NLRP3 Inflammasome Activation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:7603943. [PMID: 36785753 PMCID: PMC9922194 DOI: 10.1155/2023/7603943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/25/2022] [Accepted: 11/24/2022] [Indexed: 02/06/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease in preterm infants that may cause persistent lung injury. Artesunate exhibits excellent anti-inflammatory in lung injury caused by various factors. This study aimed to investigate the effect of the artesunate on hyperoxia-induced lung injury in neonatal mice and its mechanism. A BPD model of hyperoxic lung injury in neonatal mice was established after hyperoxia (75% oxygen) exposure for 14 days, and part of the mice received intraperitoneal injections of the artesunate. H&E staining was used to observe the pathology of lung tissue, and the degree of oxidative stress in the lung tissue was determined by commercial kits. The levels of inflammatory cytokines in the serum and lung tissues of neonatal mice were detected by an enzyme-linked immunosorbent assay. Immunohistochemical experiments were performed to further evaluate the expression of IL-1β. The real-time quantitative polymerase chain reaction was used to determine the mRNA level of the NLRP3 inflammasome. The western blot assay was used to measure the levels of NLRP3 inflammasome and NF-κB pathway-related proteins. Artesunate ameliorated weight loss and lung tissue injury in neonatal mice induced by hyperoxia. The level of malondialdehyde was decreased, while the activity of superoxide dismutase and the level of glutathione increased after artesunate treatment. Artesunate reduced the level of inflammation cytokines TNF-α, IL-6, and IL-1β in the serum and lung. Moreover, artesunate inhibited the mRNA expression and protein levels of NLRP3, ASC, and caspase-1, as well as the phosphorylation of the NF-κB and IκBα. Our findings suggest that artesunate treatment can attenuate hyperoxia-induced lung injury in BPD neonatal mice by inhibiting the activation of NLRP3 inflammasome and the phosphorylation of the NF-κB pathway.
Collapse
|
22
|
Abstract
Bronchopulmonary dysplasia (BPD) in neonates is the most common pulmonary disease that causes neonatal mortality, has complex pathogenesis, and lacks effective treatment. It is associated with chronic obstructive pulmonary disease, pulmonary hypertension, and right ventricular hypertrophy. The occurrence and development of BPD involve various factors, of which premature birth is the most crucial reason for BPD. Under the premise of abnormal lung structure and functional product, newborns are susceptible to damage to oxides, free radicals, hypoxia, infections and so on. The most influential is oxidative stress, which induces cell death in different ways when the oxidative stress balance in the body is disrupted. Increasing evidence has shown that programmed cell death (PCD), including apoptosis, necrosis, autophagy, and ferroptosis, plays a significant role in the molecular and biological mechanisms of BPD and the further development of the disease. Understanding the mode of PCD and its signaling pathways can provide new therapeutic approaches and targets for the clinical treatment of BPD. This review elucidates the mechanism of BPD, focusing on the multiple types of PCD in BPD and their molecular mechanisms, which are mainly based on experimental results obtained in rodents.
Collapse
|
23
|
Sonny S, Yuan H, Chen S, Duncan MR, Chen P, Benny M, Young K, Park KK, Schmidt AF, Wu S. GSDMD deficiency ameliorates hyperoxia-induced BPD and ROP in neonatal mice. Sci Rep 2023; 13:143. [PMID: 36599874 DOI: 10.1038/s41598-022-27201-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are among the most common morbidities affecting extremely premature infants who receive oxygen therapy. Many clinical studies indicate that BPD is associated with advanced ROP. However, the mechanistic link between hyperoxia, BPD, and ROP remains to be explored. Gasdermin D (GSDMD) is a key executor of inflammasome-induced pyroptosis and inflammation. Inhibition of GSDMD has been shown to attenuate hyperoxia-induced BPD and brain injury in neonatal mice. The objective of this study was to further define the mechanistic roles of GSDMD in the pathogenesis of hyperoxia-induced BPD and ROP in mouse models. Here we show that global GSDMD knockout (GSDMD-KO) protects against hyperoxia-induced BPD by reducing macrophage infiltration, improving alveolarization and vascular development, and decreasing cell death. In addition, GSDMD deficiency prevented hyperoxia-induced ROP by reducing vasoobliteration and neovascularization, improving thinning of multiple retinal tissue layers, and decreasing microglial activation. RNA sequencing analyses of lungs and retinas showed that similar genes, including those from inflammatory, cell death, tissue remodeling, and tissue and vascular developmental signaling pathways, were induced by hyperoxia and impacted by GSDMD-KO in both models. These data highlight the importance of GSDMD in the pathogenesis of BPD and ROP and suggest that targeting GSDMD may be beneficial in preventing and treating BPD and ROP in premature infants.
Collapse
Affiliation(s)
- Sarah Sonny
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Huijun Yuan
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Shaoyi Chen
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Matthew R Duncan
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Pingping Chen
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Merline Benny
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Karen Young
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miami, FL, USA
| | - Augusto F Schmidt
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Shu Wu
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA.
| |
Collapse
|
24
|
Wang X, Lv S, Sun J, Zhang M, Zhang L, Sun Y, Zhao Z, Wang D, Zhao X, Zhang J. Caffeine reduces oxidative stress to protect against hyperoxia-induced lung injury via the adenosine A2A receptor/cAMP/PKA/Src/ERK1/2/p38MAPK pathway. Redox Rep 2022; 27:270-278. [DOI: 10.1080/13510002.2022.2143114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Xijuan Wang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Shuai Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jianwei Sun
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Meihui Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Lei Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Yan Sun
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Ziyan Zhao
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Dandan Wang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Xinjing Zhao
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Jiajie Zhang
- Department of Paediatrics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
25
|
Fan FS. Coffee reduces the risk of hepatocellular carcinoma probably through inhibition of NLRP3 inflammasome activation by caffeine. Front Oncol 2022; 12:1029491. [PMID: 36330474 PMCID: PMC9623052 DOI: 10.3389/fonc.2022.1029491] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/03/2022] [Indexed: 11/20/2022] Open
|
26
|
Fan FS. Inhibition of NLRP3 inflammasome activation by caffeine might be a potential mechanism to reduce the risk of squamous cell carcinoma of the oral cavity and oropharynx with coffee drinking. FRONTIERS IN ORAL HEALTH 2022; 3:1017543. [PMID: 36325196 PMCID: PMC9619210 DOI: 10.3389/froh.2022.1017543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/07/2022] [Indexed: 11/07/2022] Open
|
27
|
Alva R, Mirza M, Baiton A, Lazuran L, Samokysh L, Bobinski A, Cowan C, Jaimon A, Obioru D, Al Makhoul T, Stuart JA. Oxygen toxicity: cellular mechanisms in normobaric hyperoxia. Cell Biol Toxicol 2022; 39:111-143. [PMID: 36112262 PMCID: PMC9483325 DOI: 10.1007/s10565-022-09773-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/07/2022] [Indexed: 12/15/2022]
Abstract
In clinical settings, oxygen therapy is administered to preterm neonates and to adults with acute and chronic conditions such as COVID-19, pulmonary fibrosis, sepsis, cardiac arrest, carbon monoxide poisoning, and acute heart failure. In non-clinical settings, divers and astronauts may also receive supplemental oxygen. In addition, under current standard cell culture practices, cells are maintained in atmospheric oxygen, which is several times higher than what most cells experience in vivo. In all the above scenarios, the elevated oxygen levels (hyperoxia) can lead to increased production of reactive oxygen species from mitochondria, NADPH oxidases, and other sources. This can cause cell dysfunction or death. Acute hyperoxia injury impairs various cellular functions, manifesting ultimately as physiological deficits. Chronic hyperoxia, particularly in the neonate, can disrupt development, leading to permanent deficiencies. In this review, we discuss the cellular activities and pathways affected by hyperoxia, as well as strategies that have been developed to ameliorate injury.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Maha Mirza
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Adam Baiton
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lucas Lazuran
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Lyuda Samokysh
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ava Bobinski
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Cale Cowan
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Alvin Jaimon
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Dede Obioru
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Tala Al Makhoul
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
28
|
CREB1 Transcriptionally Activates LTBR to Promote the NF-κB Pathway and Apoptosis in Lung Epithelial Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9588740. [PMID: 36118831 PMCID: PMC9481394 DOI: 10.1155/2022/9588740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a prevalent chronic pediatric lung disease. Aberrant proliferation and apoptosis of lung epithelial cells are important in the pathogenesis of BPD. Lymphotoxin beta receptor (LTBR) is expressed in lung epithelial cells. Blocking LTBR induces regeneration of lung tissue and reverts airway fibrosis in young and aged mice. This study is aimed at revealing the role of LTBR in BPD. A mouse model of BPD and two in vitro models of BPD using A549 cells and type II alveolar epithelial (ATII) cells were established by exposure to hyperoxia. We found that LTBR and CREB1 exhibited a significant upregulation in lungs of mouse model of BPD. LTBR and CREB1 expression were also increased by hyperoxia in A549 and ATII cells. According to results of cell counting kit-8 assay and flow cytometry analysis, silencing of LTBR rescued the suppressive effect of hyperoxia on cell viability and its promotive effect on cell apoptosis of A549 and ATII cells. Bioinformatics revealed CREB1 as a transcriptional factor for LTBR, and the luciferase reporter assay and ChIP assay subsequently confirmed it. The NF-κB pathway was regulated by LTBR. CREB1 induced LTBR expression at the transcriptional level to regulate NF-κB pathway and further modulate A549 and ATII cells viability and apoptosis. In conclusion, this study revealed the CREB1/LTBR/NF-κB pathway in BPD and supported the beneficial role of LTBR silence in BPD by promoting viability and decreasing apoptosis of lung epithelial cells.
Collapse
|
29
|
Caffeine Inhibits NLRP3 Inflammasome Activation by Downregulating TLR4/MAPK/NF-κB Signaling Pathway in an Experimental NASH Model. Int J Mol Sci 2022; 23:ijms23179954. [PMID: 36077357 PMCID: PMC9456282 DOI: 10.3390/ijms23179954] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/13/2022] Open
Abstract
Caffeine elicits protective effects against liver diseases, such as NASH; however, its mechanism of action involving the pyrin domain-containing-3 (NLRP3) inflammasome signaling pathway remains to be elucidated. This study aimed to evaluate the effect of caffeine on the NLRP3 inflammasome signaling pathway in a rat model of NASH. NASH was induced by feeding rats a high-fat, -sucrose, and -cholesterol diet (HFSCD) for 15 weeks along with a weekly low dose (400 mg/kg, i.p.) of CCl4. Caffeine was administered at 50 mg/kg p.o. The effects of HFSCD+CCl4 and caffeine on the liver were evaluated using biochemical, ultrastructural, histological, and molecular biological approaches. The HFSCD+CCl4-treated rats showed fat accumulation in the liver, elevated levels of inflammatory mediators, NLRP3 inflammasome activation, antioxidant dysregulation, and liver fibrosis. Caffeine reduced necrosis, cholestasis, oxidative stress, and fibrosis. Caffeine exhibited anti-inflammatory effects by attenuating NLRP3 inflammasome activation. Moreover, caffeine prevented increases in toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) protein levels and mitigated the phosphorylation of mitogen-activated protein kinase (MAPK). Importantly, caffeine prevented the activation of hepatic stellate cells. This study is the first to report that caffeine ameliorates NASH by inhibiting NLRP3 inflammasome activation through the suppression of the TLR4/MAPK/NF-κB signaling pathway.
Collapse
|
30
|
Liu W, Han X, Li Q, Sun L, Wang J. Iguratimod ameliorates bleomycin-induced pulmonary fibrosis by inhibiting the EMT process and NLRP3 inflammasome activation. Biomed Pharmacother 2022; 153:113460. [DOI: 10.1016/j.biopha.2022.113460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022] Open
|
31
|
Yang X, Jiang S, Deng X, Luo Z, Chen A, Yu R. Effects of Antioxidants in Human Milk on Bronchopulmonary Dysplasia Prevention and Treatment: A Review. Front Nutr 2022; 9:924036. [PMID: 35923207 PMCID: PMC9340220 DOI: 10.3389/fnut.2022.924036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe chronic lung illness that affects neonates, particularly premature infants. It has far-reaching consequences for infant health and their families due to intractable short- and long-term repercussions. Premature infant survival and long-term quality of life are severely harmed by BPD, which is characterized by alveolarization arrest and hypoplasia of pulmonary microvascular cells. BPD can be caused by various factors, with oxidative stress (OS) being the most common. Premature infants frequently require breathing support, which results in a hyperoxic environment in the developing lung and obstructs lung growth. OS can damage the lungs of infants by inducing cell death, inhibiting alveolarization, inducing inflammation, and impairing pulmonary angiogenesis. Therefore, antioxidant therapy for BPD relieves OS and lung injury in preterm newborns. Many antioxidants have been found in human milk, including superoxide dismutase, glutathione peroxidase, glutathione, vitamins, melatonin, short-chain fatty acids, and phytochemicals. Human milk oligosaccharides, milk fat globule membrane, and lactoferrin, all unique to human milk, also have antioxidant properties. Hence, human milk may help prevent OS injury and improve BPD prognosis in premature infants. In this review, we explored the role of OS in the pathophysiology of BPD and related signaling pathways. Furthermore, we examined antioxidants in human milk and how they could play a role in BPD to understand whether human milk could prevent and treat BPD.
Collapse
Affiliation(s)
- Xianpeng Yang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xianhui Deng
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ailing Chen
- Translational Medicine Laboratory, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Ailing Chen
| | - Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Renqiang Yu
| |
Collapse
|
32
|
Yuan Y, Yang Y, Lei X, Dong W. Caffeine and bronchopulmonary dysplasia: Clinical benefits and the mechanisms involved. Pediatr Pulmonol 2022; 57:1392-1400. [PMID: 35318830 DOI: 10.1002/ppul.25898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease that occurs during the neonatal period and is commonly associated with prematurity. This condition results in a severe economic burden on society and the families involved. Caffeine is used not only for the treatment of apnea in prematurity, but also for the prevention of BPD. There are multiple clinical benefits of caffeine treatment, including improved extubation success, a reduced duration of mechanical ventilation, improved lung function, and a reduction of patent ductus arteriosus requiring treatment. These clinical benefits of caffeine for the treatment of BPD are supported by both clinical trials and evidence from animal models. However, the mechanism by which caffeine protects against BPD remains unclear. Here, we review the clinical value of caffeine in the prevention of BPD and its potential mechanisms of action, including anti-inflammatory, antioxidant, antifibrotic, and antiapoptotic properties, the regulation of angiogenesis, and diuretic effects. Our aim is to provide a new theoretical basis for the clinical treatment of BPD.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Yang
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoping Lei
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
33
|
Wang X, Huo R, Liang Z, Xu C, Chen T, Lin J, Li L, Lin W, Pan B, Fu X, Chen S. Simvastatin Inhibits NLRP3 Inflammasome Activation and Ameliorates Lung Injury in Hyperoxia-Induced Bronchopulmonary Dysplasia via the KLF2-Mediated Mechanism. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8336070. [PMID: 35509841 PMCID: PMC9060986 DOI: 10.1155/2022/8336070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly found in premature infants. Excessive inflammation and oxidative stress contribute to BPD occurrence and development. Simvastatin, as an inhibitor of HMG-CoA reductase, has been reported to have antioxidative and anti-inflammatory effects. However, its effect and possible mechanisms in hyperoxia-induced lung injury are rarely reported. In this study, in vivo and in vitro experiments were conducted to investigate whether simvastatin could ameliorate hyperoxia-induced lung injury and explore its potential mechanism. For the in vivo study, simvastatin could improve alveolar development after hyperoxic lung injury and reduce hyperoxic stress and inflammation. The in vitro study revealed that simvastatin can reduce inflammation in A549 cells after high-oxygen exposure. Simvastatin suppressed NLRP3 inflammasome activation and played anti-inflammatory and antioxidant roles by increasing KLF2 (Krüppel-like factor 2) expression. In vitro experiments also revealed that these effects of simvastatin were partially reversed by KLF2 shRNA, indicating that KLF2 was involved in simvastatin effects. In summary, our findings indicate that simvastatin could downregulate NLRP3 inflammasome activation and attenuate lung injury in hyperoxia-induced bronchopulmonary dysplasia via KLF2-mediated mechanism.
Collapse
Affiliation(s)
- Xinye Wang
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Department of Pediatric, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ran Huo
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Zhongjie Liang
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Congcong Xu
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Tingting Chen
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jingjing Lin
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Luyao Li
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Wei Lin
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Bingting Pan
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoqin Fu
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shangqin Chen
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
34
|
Gilfillan M, Bhandari V. Moving Bronchopulmonary Dysplasia Research from the Bedside to the Bench. Am J Physiol Lung Cell Mol Physiol 2022; 322:L804-L821. [PMID: 35437999 DOI: 10.1152/ajplung.00452.2021] [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: 11/22/2022] Open
Abstract
Although advances in the respiratory management of extremely preterm infants have led to improvements in survival, this progress has not yet extended to a reduction in the incidence of bronchopulmonary dysplasia (BPD). BPD is a complex multifactorial condition that primarily occurs due to disturbances in the regulation of normal pulmonary airspace and vascular development. Preterm birth and exposure to invasive mechanical ventilation also compromises large airway development, leading to significant morbidity and mortality. Although both predisposing and protective genetic and environmental factors have been frequently described in the clinical literature, these findings have had limited impact on the development of effective therapeutic strategies. This gap is likely because the molecular pathways that underlie these observations are yet not fully understood, limiting the ability of researchers to identify novel treatments that can preserve normal lung development and/or enhance cellular repair mechanisms. In this review article, we will outline various well-established clinical observations whilst identifying key knowledge gaps that need to be filled with carefully designed pre-clinical experiments. We will address these issues by discussing controversial topics in the pathophysiology, the pathology and the treatment of BPD, including an evaluation of existing animal models that have been used to answer important questions.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Division of Neonatology, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA
| | - Vineet Bhandari
- Division of Neonatology, The Children's Regional Hospital at Cooper/Cooper Medical School of Rowan University, Camden, NJ
| |
Collapse
|
35
|
Zhang S, Luan X, Li H, Jin Z. Insulin-like growth factor-1: A potential target for bronchopulmonary dysplasia treatment (Review). Exp Ther Med 2022; 23:191. [PMID: 35126694 PMCID: PMC8794548 DOI: 10.3892/etm.2022.11114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/08/2021] [Indexed: 11/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common respiratory disorder among preterm infants, particularly low-birth-weight infants (LBWIs) and very-low-birth-weight infants (VLBWIs). Although BPD was first reported 50 years ago, no specific drugs or efficient measures are yet available for prevention or treatment. Insulin-like growth factor-1 (IGF-1) belongs to the insulin family. It promotes mitosis and stimulates cell proliferation and DNA synthesis, the primary factors involved in pulmonary development during the fetal and postnatal periods. Several studies have reported that IGF-1 exerts certain effects on BPD genesis and progression by regulating BPD-related biological processes. In addition, exogenous addition of IGF-1 can alleviate lung inflammation, cell apoptosis and eliminate alveolar development disorders in children with BPD. These findings suggest that IGF-1 could be a new target for treating BPD. Here, we summarize and analyze the definition, pathogenesis, and research status of BPD, as well as the pathogenesis of IGF-1 in BPD and the latest findings in related biological processes.
Collapse
Affiliation(s)
- Shujian Zhang
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Xue Luan
- Department of Pediatrics, First Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Huiwen Li
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
36
|
Qing C, Ziyun L, Xuefei Y, Xinyi Z, Xindong X, Jianhua F. Protective Effects of 18β-Glycyrrhetinic Acid on Neonatal Rats with Hyperoxia Exposure. Inflammation 2022; 45:1224-1238. [PMID: 34989920 DOI: 10.1007/s10753-021-01616-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/05/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common devastating pulmonary complication in preterm infants. Supplemental oxygen is a lifesaving therapeutic measure used for premature infants with pulmonary insufficiency. However, oxygen toxicity is a significant trigger for BPD. Oxidative stress disrupts lung development, accompanied by increased pro-inflammatory cytokines and chemokines expression and immune cells infiltration in lung tissue. Licorice, a typical traditional herbal medicine, is commonly used in the medicine and food industries. 18β-Glycyrrhetinic acid (18β-GA), a primary active ingredient of licorice, has powerful anti-oxidative and anti-inflammatory effects. This study aimed to determine whether 18β-GA has a protective effect on neonatal rats with hyperoxia exposure. Newborn Sprague-Dawley rats were kept in either 21% (normoxia) or 80% O2 (hyperoxia) continuously from postnatal day (PN) 1 to 14. 18β-GA was injected intragastrically at 50 or 100 mg/kg body weight once a day from PN 1 to 14. We examined the body weight and alveolar development and measured ROS level and the markers of pulmonary inflammation. Mature-IL-1β and NF-κB pathway proteins, and the NLRP3 inflammasome, were assessed; concurrently, caspase-1 activity was measured. Our results indicated that hyperoxia resulted in alveolar simplification and decreased bodyweight of neonatal rats. Hyperoxia increased ROS level and pulmonary inflammation and activated NF-κB and the NLRP3 inflammasome. 18β-GA treatment inhibited the activation of NF-κB and the NLRP3 inflammasome, decreased ROS level and pulmonary inflammation, improved alveolar development, and increased the bodyweight of neonatal rats with hyperoxia exposure. Our study demonstrates that 18β-GA has a protective effect on neonatal rats with hyperoxia exposure.
Collapse
Affiliation(s)
- Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Liu Ziyun
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
37
|
Tian C, Li D, Fu J. Molecular Mechanism of Caffeine in Preventing Bronchopulmonary Dysplasia in Premature Infants. Front Pediatr 2022; 10:902437. [PMID: 35795332 PMCID: PMC9251307 DOI: 10.3389/fped.2022.902437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory complication commonly seen in premature infants. Following continuous advances in neonatal intensive care diagnosis and treatment technology, an increasing number of premature babies are being treated successfully. Despite these remarkable improvements, there has been no significant decline in the incidence of BPD; in fact, its incidence has increased as more extremely preterm infants survive. Therefore, in view of the impact of BPD on the physical and mental health of children and the increased familial and social burden on these children, early prevention of BPD is emphasized. In recent decades, the clinical application of caffeine in treating primary apnea in premature infants was shown not only to stimulate the respiratory center but also to confer obvious protection to the nervous and respiratory systems. Numerous clinical cross-sectional and longitudinal studies have shown that caffeine plays a significant role in the prevention and treatment of BPD, but there is a lack of overall understanding of its potential molecular mechanisms. In this review, we summarize the possible molecular mechanisms of caffeine in the prevention or treatment of BPD, aiming to better guide its clinical application.
Collapse
Affiliation(s)
- Congliang Tian
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.,Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
38
|
Sherif IO, Al-Shaalan NH. OUP accepted manuscript. Toxicol Res (Camb) 2022; 11:417-425. [PMID: 35782645 PMCID: PMC9244212 DOI: 10.1093/toxres/tfac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 11/14/2022] Open
Abstract
Acute lung injury has been reported following various chemotherapeutic agents administration. Several pathways for lung injury have been speculated however, the exact mechanism of the lung injury induced by methotrexate (MTX) is yet to be defined. The potential protective effect of Ginkgo biloba extract (GB), a Chinese herbal medicine, against MTX-induced lung injury is still not reported. Therefore, this study was performed to examine the possible implication of NLRP3 inflammasome and miRNA-21 in the pathogenesis of the MTX-induced lung injury as well as the protective role of GB in ameliorating the induced lung injury. Rats received GB (100 mg/kg/day, orally) for 10 days and MTX (20 mg/kg single dose, intraperitoneally) on the fifth day. MTX-induced lung injury was manifested by lung histopathology. MTX exhibited a marked increase in lung malondialdehyde beside a notable decrease in lung reduced glutathione. Moreover, MTX injection activated the lung NLRP3 inflammasome by significant upregulation of the NLRP3, ASC, caspase-1 lung mRNA expressions and protein levels in addition to lung NF-kBp65 protein expression, and miRNA-21 expression when compared with the normal control group. However, GB administration mitigated lung injury and inhibited the NLRP3 activation. This study is the first report to reveal the involvement of NLRP3 inflammasome in the pathogenesis of MTX-induced lung injury and also to show that the administration of GB alleviates the lung injury induced by MTX via suppressing the oxidative stress, restoring the antioxidant activity, blocking the NLRP3/ASC/Caspase-1 signaling and downregulating the NF-kBp65 protein expression ae well as miRNA-21 expression in lung tissue.
Collapse
Affiliation(s)
- Iman O Sherif
- Corresponding Author: Dr. Iman O. Sherif, PhD, Consultant of Biochemistry, Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt, ;
| | - Nora H Al-Shaalan
- Chemistry Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| |
Collapse
|
39
|
Vyas-Read S, Logan JW, Cuna AC, Machry J, Leeman KT, Rose RS, Mikhael M, Wymore E, Ibrahim JW, DiGeronimo RJ, Yallapragada S, Haberman BE, Padula MA, Porta NF, Murthy K, Nelin LD, Coghill CH, Zaniletti I, Savani RC, Truog W, Engle WA, Lagatta JM. A comparison of newer classifications of bronchopulmonary dysplasia: findings from the Children's Hospitals Neonatal Consortium Severe BPD Group. J Perinatol 2022; 42:58-64. [PMID: 34354227 PMCID: PMC8340076 DOI: 10.1038/s41372-021-01178-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/08/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To compare three bronchopulmonary dysplasia (BPD) definitions against hospital outcomes in a referral-based population. STUDY DESIGN Data from the Children's Hospitals Neonatal Consortium were classified by 2018 NICHD, 2019 NRN, and Canadian Neonatal Network (CNN) BPD definitions. Multivariable models evaluated the associations between BPD severity and death, tracheostomy, or length of stay, relative to No BPD references. RESULTS Mortality was highest in 2019 NRN Grade 3 infants (aOR 225), followed by 2018 NICHD Grade 3 (aOR 145). Infants with lower BPD grades rarely died (<1%), but Grade 2 infants had aOR 7-21-fold higher for death and 23-56-fold higher for tracheostomy. CONCLUSIONS Definitions with 3 BPD grades had better discrimination and Grade 3 2019 NRN had the strongest association with outcomes. No/Grade 1 infants rarely had severe outcomes, but Grade 2 infants were at risk. These data may be useful for counseling families and determining therapies for infants with BPD.
Collapse
Affiliation(s)
- Shilpa Vyas-Read
- Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - J. Wells Logan
- grid.261331.40000 0001 2285 7943The Ohio State University and Nationwide Children’s Hospital, Columbus, OH USA
| | - Alain C. Cuna
- grid.266756.60000 0001 2179 926XUniversity of Missouri-Kansas City and Children’s Mercy Hospital, Kansas, MO USA
| | - Joana Machry
- grid.21107.350000 0001 2171 9311Johns Hopkins University and Johns Hopkins All Children’s Hospital, Baltimore, MD USA
| | - Kristin T. Leeman
- grid.2515.30000 0004 0378 8438Harvard University and Boston Children’s Hospital, Cambridge, MA USA
| | - Rebecca S. Rose
- grid.257410.50000 0004 0413 3089Indiana University and Riley Children’s Hospital, Bloomington, IN USA
| | - Michel Mikhael
- grid.19006.3e0000 0000 9632 6718University of California, Irvine and Children’s Hospital of Orange County, Los Angeles, CA USA
| | - Erica Wymore
- grid.266190.a0000000096214564University of Colorado and Children’s Hospital Colorado, Boulder, CO USA
| | - John W. Ibrahim
- grid.239553.b0000 0000 9753 0008University of Pittsburgh and Children’s Hospital of Pittsburgh, Pittsburgh, PA USA
| | - Robert J. DiGeronimo
- grid.34477.330000000122986657University of Washington, Seattle and Seattle Children’s Hospital, Seattle, WA USA
| | - Sushmita Yallapragada
- University of Texas, Southwestern and Children’s Medical Center of Dallas, Dallas, TX USA
| | - Beth E. Haberman
- grid.24827.3b0000 0001 2179 9593University of Cincinnati and Cincinnati Children’s Hospital, Cincinnati, OH USA
| | - Michael A. Padula
- grid.239552.a0000 0001 0680 8770University of Pennsylvania and Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Nicolas F. Porta
- grid.413808.60000 0004 0388 2248Northwestern University and Ann & Robert H Lurie Children’s Hospital, Evanston, IL USA
| | - Karna Murthy
- grid.413808.60000 0004 0388 2248Northwestern University and Ann & Robert H Lurie Children’s Hospital, Evanston, IL USA
| | - Leif D. Nelin
- grid.261331.40000 0001 2285 7943The Ohio State University and Nationwide Children’s Hospital, Columbus, OH USA
| | - Carl H. Coghill
- grid.265892.20000000106344187University of Alabama, Birmingham and Children’s of Alabama, Birmingham, AL USA
| | | | - Rashmin C. Savani
- University of Texas, Southwestern and Children’s Medical Center of Dallas, Dallas, TX USA
| | - William Truog
- grid.266756.60000 0001 2179 926XUniversity of Missouri-Kansas City and Children’s Mercy Hospital, Kansas, MO USA
| | - William A. Engle
- grid.257410.50000 0004 0413 3089Indiana University and Riley Children’s Hospital, Bloomington, IN USA
| | - Joanne M. Lagatta
- grid.30760.320000 0001 2111 8460Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, WI USA
| |
Collapse
|
40
|
Mechanism of Adipose-Derived Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying miR-21-5p in Hyperoxia-Induced Lung Injury. Stem Cell Rev Rep 2021; 18:1007-1024. [PMID: 34882302 DOI: 10.1007/s12015-021-10311-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2021] [Indexed: 01/10/2023]
Abstract
Hyperoxia-induced lung injury (HILI) tends to develop bronchopulmonary dysplasia. Adipose-derived mesenchymal stem cell (ADMSC)-derived extracellular vesicles (EVs) hold great promise in alleviating lung injury. This study explored the mechanism of ADMSC-EVs in HILI. ADMSC-EVs were isolated and identified. The murine and cell models of HILI were established. HILI mice and cells were pre-treated with ADMSC-EVs. The lung dry/wet ratio, pathological structure, apoptosis, and inflammation of HILI mice were measured. The viability, apoptosis, and oxidative stress of HILI cells were measured. The internalization of EVs in lung and cells was observed by fluorescence labeling. The binding relationships between miR-21-5p and SKP2, and Nr2f2 and C/EBPα were analyzed. The binding of SKP2 and Nr2f2 and the Nr2f2 ubiquitination level were detected. ADMSC-EVs exerted preventive effects on HILI mice, evidenced by reduced lung dry/wet ratio, inflammation, and apoptosis in HILI mice. In vitro, EVs enhanced HILI cell viability and reduced apoptosis, inflammation, and oxidative stress. EVs carried miR-21-5p into lung cells to upregulate miR-21-5p expression and thereby target SKP2. SKP2 bound to Nr2f2 and promoted its ubiquitination degradation. EVs inhibited the binding of Nr2f2 and C/EBPα and further suppressed C/EBPα transcription. Collectively, ADMSC-EVs carrying miR-21-5p alleviated HILI via the SKP2/Nr2f2/C/EBPα axis. Role and mechanism of adipose-derived mesenchymal stem cell-derived extracellular vesicles in hyperoxia-induced lung injury. ADMSC-EVs upregulated miR-21-5p expression in cells by carrying miR-21-5p into lung cells, thereby promoting the binding of miR-21-5p and SKP2 mRNA, inhibiting the expression of SKP2, reducing the ubiquitination level of Nr2f2, increasing the expression of Nr2f2, promoting the binding of Nr2f2 and the C/EBPα promoter, upregulating C/EBPα mRNA level, and eventually alleviating HILI.
Collapse
|
41
|
Jia D, Zheng J, Zhou Y, Jia J, Ye X, Zhou B, Chen X, Mo Y, Wang J. Ferroptosis is Involved in Hyperoxic Lung Injury in Neonatal Rats. J Inflamm Res 2021; 14:5393-5401. [PMID: 34703276 PMCID: PMC8536887 DOI: 10.2147/jir.s335061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/11/2021] [Indexed: 12/20/2022] Open
Abstract
Purpose To evaluate whether ferroptosis is involved in hyperoxic acute lung injury (HALI) and its mechanisms through the HALI model. Methods HE staining was used to assess lung injury pathology after the establishment of neonatal rat HALI model. ELISA was used to detect ROS, GPX4, and GSH expression. Prussian blue staining and Western Blot were used to detect iron deposition and the expression of ferroptosis-related proteins, respectively. Results The HALI group showed pathological changes with larger and fewer alveoli and thicker alveolar septa after HE staining. Prussian blue staining detected significant iron deposition in the lung tissue of the HALI group. GPX4, GSH, GSS, and SLC7A11 expressions were significantly decreased in the HALI group than in the normal control group. In contrast, ROS, TFRC, FHC, and FLC expressions showed opposite results (p<0.05). Conclusion Ferroptosis may be involved in the pathological process of hyperoxic lung injury in neonatal rats.
Collapse
Affiliation(s)
- Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jinqiu Jia
- Department of Pediatric, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, 317599, Zhejiang, People's Republic of China
| | - Xiaoxiao Ye
- Department of Nursing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Bingbing Zhou
- Department of Nursing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Xingxing Chen
- Department of Nursing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yunchang Mo
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| |
Collapse
|
42
|
Oxygen Toxicity to the Immature Lung-Part I: Pathomechanistic Understanding and Preclinical Perspectives. Int J Mol Sci 2021; 22:ijms222011006. [PMID: 34681665 PMCID: PMC8540649 DOI: 10.3390/ijms222011006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/13/2023] Open
Abstract
In utero, the fetus and its lungs develop in a hypoxic environment, where HIF-1α and VEGFA signaling constitute major determinants of further development. Disruption of this homeostasis after preterm delivery and extrauterine exposure to high fractions of oxygen are among the key events leading to bronchopulmonary dysplasia (BPD). Reactive oxygen species (ROS) production constitutes the initial driver of pulmonary inflammation and cell death, altered gene expression, and vasoconstriction, leading to the distortion of further lung development. From preclinical studies mainly performed on rodents over the past two decades, the deleterious effects of oxygen toxicity and the injurious insults and downstream cascades arising from ROS production are well recognized. This article provides a concise overview of disease drivers and different therapeutic approaches that have been successfully tested within experimental models. Despite current studies, clinical researchers are still faced with an unmet clinical need, and many of these strategies have not proven to be equally effective in clinical trials. In light of this challenge, adapting experimental models to the complexity of the clinical situation and pursuing new directions constitute appropriate actions to overcome this dilemma. Our review intends to stimulate research activities towards the understanding of an important issue of immature lung injury.
Collapse
|
43
|
Ozdemir R, Gokce IK, Taslidere AC, Tanbek K, Gul CC, Sandal S, Turgut H, Kaya H, Aslan M. Does Chrysin prevent severe lung damage in Hyperoxia-Induced lung injury Model? Int Immunopharmacol 2021; 99:108033. [PMID: 34343938 DOI: 10.1016/j.intimp.2021.108033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Oxidative stress and inflammation play a critical role in the etiopathogenesis of bronchopulmonary dysplasia (BPD). The aim of this study was to evaluate the preventive effect of Chrysin (CH), an antioxidant, antiinflammatory, antiapoptotic and antifibrotic drug, on hyperoxia-induced lung injury in a neonatal rat model. METHODS Forty infant rats were divided into four groups labeled the Control, CH, BPD, and BPD + CH. The control and CH groups were kept in a normal room environment, while the BPD and BPD + CH groups were kept in a hyperoxic (90-95%) environment. At the end of the study, lung tissue was evaluated with respect to apoptosis, histopathological damage and alveolar macrophage score as well as oxidant capacity, antioxidant capacity, and inflammation. RESULTS Compared to the BPD + CH and control groups, the lung tissues of the BPD group displayed substantially higher levels of MDA, TOS, TNF-α, and IL-1β (p < 0.05). While the BPD + CH group showed similar levels of TNF-α and IL-1β as the control group, MDA and TOS levels were higher than the control group, and significantly lower than the BPD group (p < 0.05). The BPD group exhibited considerably lower levels of TAS, SOD, GSH, and GSH-Px in comparison to the control group (p < 0.05). The BPD and BPD + CH groups exhibited higher mean scores of histopathological damage and alveolar macrophage when compared to the control and CH groups (p ≤ 0.0001). Both scores were found to be lower in the BPD + CH group in comparison to the BPD group (p ≤ 0.0001). The BPD + CH group demonstrated a significantly lower average of TUNEL and caspase-3 positive cells than the BPD group. CONCLUSION We found that prophylaxis with CH results in lower histopathological damage score and reduces apoptotic cell count, inflammation and oxidative stress while increasing anti-oxidant capacity.
Collapse
Affiliation(s)
- Ramazan Ozdemir
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey.
| | - Ismail Kursat Gokce
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Asli Cetin Taslidere
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Kevser Tanbek
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Cemile Ceren Gul
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Suleyman Sandal
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Turgut
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Huseyin Kaya
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Aslan
- Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| |
Collapse
|
44
|
Wickramasinghe LC, van Wijngaarden P, Tsantikos E, Hibbs ML. The immunological link between neonatal lung and eye disease. Clin Transl Immunology 2021; 10:e1322. [PMID: 34466225 PMCID: PMC8387470 DOI: 10.1002/cti2.1322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/02/2021] [Accepted: 07/13/2021] [Indexed: 01/02/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are two neonatal diseases of major clinical importance, arising in large part as a consequence of supplemental oxygen therapy used to promote the survival of preterm infants. The presence of coincident inflammation in the lungs and eyes of neonates receiving oxygen therapy indicates that a dysregulated immune response serves as a potential common pathogenic factor for both diseases. This review examines the current state of knowledge of immunological dysregulation in BPD and ROP, identifying similarities in the cellular subsets and inflammatory cytokines that are found in the alveoli and retina during the active phase of these diseases, indicating possible mechanistic overlap. In addition, we highlight gaps in the understanding of whether these responses emerge independently in the lung and retina as a consequence of oxygen exposure or arise because of inflammatory spill-over from the lung. As BPD and ROP are anatomically distinct, they are often considered discreet disease entities and are therefore treated separately. We propose that an improved understanding of the relationship between BPD and ROP is key to the identification of novel therapeutic targets to treat or prevent both conditions simultaneously.
Collapse
Affiliation(s)
- Lakshanie C Wickramasinghe
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Peter van Wijngaarden
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVICAustralia
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVICAustralia
| | - Evelyn Tsantikos
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Margaret L Hibbs
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| |
Collapse
|
45
|
Liu B, He R, Zhang L, Hao B, Jiang W, Wang W, Geng Q. Inflammatory Caspases Drive Pyroptosis in Acute Lung Injury. Front Pharmacol 2021; 12:631256. [PMID: 33613295 PMCID: PMC7892432 DOI: 10.3389/fphar.2021.631256] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
Acute lung injury (ALI), a critical respiratory disorder that causes diffuse alveolar injury leads to high mortality rates with no effective treatment. ALI is characterized by varying degrees of ventilation/perfusion mismatch, severe hypoxemia, and poor pulmonary compliance. The diffuse injury to cells is one of most important pathological characteristics of ALI. Pyroptosis is a form of programmed cell death distinguished from apoptosis induced by inflammatory caspases, which can release inflammatory cytokines to clear cells infected by pathogens and promote monocytes to reassemble at the site of injury. And pyroptosis not only promotes inflammation in certain cell types, but also regulates many downstream pathways to perform different functions. There is increasing evidence that pyroptosis and its related inflammatory caspases play an important role in the development of acute lung injury. The main modes of activation of pyroptosis is not consistent among different types of cells in lung tissue. Meanwhile, inhibition of inflammasome, the key to initiating pyroptosis is currently the main way to treat acute lung injury. The review summarizes the relationship among inflammatory caspases, pyroptosis and acute lung injury and provides general directions and strategies to conduct further research.
Collapse
Affiliation(s)
- Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Hao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Chen CM, Hwang J, Chou HC, Chen C. Anti-Tn Monoclonal Antibody Attenuates Hyperoxia-Induced Lung Injury by Inhibiting Oxidative Stress and Inflammation in Neonatal Mice. Front Pharmacol 2020; 11:568502. [PMID: 33013407 PMCID: PMC7506044 DOI: 10.3389/fphar.2020.568502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Maternal immunization with Tn vaccine increases serum anti-Tn antibody titers and attenuates hyperoxia-induced lung injury in neonatal rats. This study determined whether anti-Tn monoclonal antibody can protect against hyperoxia-induced lung injury in neonatal mice. Newborn BALB/c mice were exposed to room air (RA) or normobaric hyperoxia (85% O2) for 1 week, creating four study groups as follows: RA + phosphate-buffered saline (PBS), RA + anti-Tn monoclonal antibody, O2 + PBS, and O2 + anti-Tn monoclonal antibody. The anti-Tn monoclonal antibody at 25 μg/g body weight in 50 μl PBS was intraperitoneally injected on postnatal days 2, 4, and 6. Hyperoxia reduced body weight and survival rate, increased mean linear intercept (MLI) and lung tumor necrosis factor-α, and decreased vascular endothelial growth factor (VEGF) expression and vascular density on postnatal day 7. Anti-Tn monoclonal antibody increased neonatal serum anti-Tn antibody titers, reduced MLI and cytokine, and increased VEGF expression and vascular density to normoxic levels. The attenuation of lung injury was accompanied by a reduction in lung oxidative stress and nuclear factor-κB activity. Anti-Tn monoclonal antibody improves alveolarization and angiogenesis in hyperoxia-injured newborn mice lungs 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
| | - Chinde Chen
- Department of Research and Development, Taivital Biopharmaceutical Co. LTD, Taoyuan, Taiwan
| |
Collapse
|
47
|
Zhang Q, Ran X, He Y, Ai Q, Shi Y. Acetate Downregulates the Activation of NLRP3 Inflammasomes and Attenuates Lung Injury in Neonatal Mice With Bronchopulmonary Dysplasia. Front Pediatr 2020; 8:595157. [PMID: 33614540 PMCID: PMC7889800 DOI: 10.3389/fped.2020.595157] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Bronchopulmonary dysplasia (BPD) is a common pulmonary complication in preterm infants. Acetate is a metabolite produced by the gut microbiota, and its anti-inflammatory function is well known. The role of acetate in BPD has not been studied. Here, we investigate the effects of acetate on lung inflammation and damage in mice model of BPD. Objective: To investigate the role of acetate in the development of BPD. Methods: C57BL/6 mice were randomly divided into three groups on the 3rd day after birth: room air group, hyperoxia group, and hyperoxia + acetate (250 mM, 0.02 ml/g) group. The expression of inflammatory factors was determined by ELISA and RT-PCR, and NLRP3 and caspase-1 were detected by Western blot. High-throughput sequencing was used to detect bacterial communities in the mice intestines. Results: After acetate treatment, the expression levels of TNF-α, IL-1β, IL-18, NLRP3, and caspase-1 were significantly reduced, while the expression of GPR43 was increased. In the BPD mice treated with acetate, the proportion of Escherichia-Shigella was lower than in placebo-treated BPD mice, while the abundance of Ruminococcus was increased. Conclusions: These results indicate that acetate may regulate intestinal flora and reduce inflammatory reactions and lung injury in BPD. Therefore, acetate may be an effective drug to protect against neonatal BPD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Xiao Ran
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yu He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Qing Ai
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| |
Collapse
|