1
|
Misar PL, Otari KV. Investigation of the protective effect of cilostazol on acute lung injury-mediated inflammation and in silico molecular modelling studies of inflammatory signalling pathway: a repurposing study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03734-z. [PMID: 39754679 DOI: 10.1007/s00210-024-03734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Acute lung injury i.e. ALI and its serious form acute respiratory distress syndrome (ARDS) are incurable medical conditions associated with significant global mortality and morbidity. The objective of the present research was to repurpose cilostazol, an antiplatelet drug with anti-inflammatory, antioxidant and antiapoptotic effect, as a potential approach for treatment of ALI. Its multifaceted effects make it promising candidate but its mechanism against ALI remains elusive. Hence it is needed to elucidate its mechanism of action to revealed its therapeutic potential and improve its clinical outcomes. This study investigated the potential inflammatory therapeutic targets of cilostazol with its protective effect against lipopolysaccharide (LPS)-induced ALI. We have identified 10 inflammatory target proteins of cilostazol i.e. PDK1, RAC1, PTK6, KDR, EGFR, endothelin-I, caspase-3, TNF-α, NF-κB1/BTK, a TLR/IRAK4 by molecular docking and validated by in vivo evaluation to demonstrate its therapeutic efficacy. In vivo experiment was performed in two sets; first to determine cellular inflammation by analysing the biomarkers in both lung homogenate and bronchoalveolar fluid and second set to study lung edema with dexamethasone as a standard. Additionally, respiratory parameters, related mRNA expressions and histopathology was evaluated. Our results, molecular docking showed that cilostazol binds to identified inflammatory target proteins with the same binding affinity as that of experimental inhibitors. In vivo, downregulated oxidative stress, and inflammation i.e. attenuated the pulmonary edema and vascular leakage, release of inflammatory mediators i.e. IL-6, TNF-α, NO, C-reactive protein (CRP), lactate dehydrogenase (LDH) myeloperoxidase (MPO), Krebs von den Lungen 6 (KL-6), and the recruitment of inflammatory cells; downregulated the m-RNA gene expressions of tumour necrosis factor alpha (TNF-α), nuclear factor kappa B( NF-kB), Toll-like receptor 4 (TLR4), Janus kinase/signal transducer, and activator of transcription 3 (JAK and STAT3); and improved total lung capacity in LPS-challenged rats. These findings revealed the cilostazol's efficacy as promising therapeutic agent for ALI by inhibiting the NF-κB/TLR4/JAK-STAT3 signalling pathway.
Collapse
Affiliation(s)
- Pranaya L Misar
- Dr. Babasaheb Ambedkar Technological University, Lonere, Raigad, 402103, India.
- N.N. Sattha College of Pharmacy, Ahmednagar, Maharashtra, 414003, India.
| | - Kishor V Otari
- Dr. Babasaheb Ambedkar Technological University, Lonere, Raigad, 402103, India
- Navsahyadri College of Pharmacy, Nasrapur, Pune, Maharashtra, 412213, India
| |
Collapse
|
2
|
Yang Y, Zhang T, Li Q, Ling Y, Ma Y, Tao S. SQSTM1 improves acute lung injury via inhibiting airway epithelium ferroptosis in a vitamin D receptor/autophagy-mediated manner. Free Radic Biol Med 2024; 222:588-600. [PMID: 38996820 DOI: 10.1016/j.freeradbiomed.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Emerging evidence has reported that acute lung injury (ALI), characterized by inflammation and oxidative stress in airway epithelium, is regulated by programmed cell death. Ferroptosis, a regulated form of cell death spurred by uncontrolled lipid peroxidation, has been proven to implicate various diseases. Inhibiting ferroptosis represents a feasible strategy for ALI through the suppression of lipid peroxidation, while the mechanism remains to be further elucidated. Here, we identified Sequestosome 1 (SQSTM1) as a negative regulator of airway epithelium ferroptosis during ALI. SQSTM1 knockdown cells manifested higher sensitivity to ferroptosis. Mechanistically, SQSTM1 was found to directly interact with vitamin D receptor (VDR) through its nuclear receptor (NR) box motif, facilitating its nuclear translocation and initiating autophagy at the transcriptional level. To further validate these findings, an in vivo preventive model utilizing spermidine, a proven inducer of SQSTM1 was established. The results consistently demonstrated that spermidine supplementation significantly induced SQSTM1 and ameliorated ALI by mitigating airway epithelial ferroptosis. Notably, these effects were abrogated in the absence of SQSTM1. Taken together, this study identified SQSTM1 as a negative regulator of airway epithelium ferroptosis in a VDR-mediated autophagy manner, making it a potential therapeutic target for the treatment of ALI.
Collapse
Affiliation(s)
- Youjing Yang
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China.
| | - Tao Zhang
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Qianmin Li
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yi Ling
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yu Ma
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Shasha Tao
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China.
| |
Collapse
|
3
|
Kutumova EO, Akberdin IR, Egorova VS, Kolesova EP, Parodi A, Pokrovsky VS, Zamyatnin, Jr AA, Kolpakov FA. Physiologically based pharmacokinetic model for predicting the biodistribution of albumin nanoparticles after induction and recovery from acute lung injury. Heliyon 2024; 10:e30962. [PMID: 38803942 PMCID: PMC11128879 DOI: 10.1016/j.heliyon.2024.e30962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/02/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The application of nanomedicine in the treatment of acute lung injury (ALI) has great potential for the development of new therapeutic strategies. To gain insight into the kinetics of nanocarrier distribution upon time-dependent changes in tissue permeability after ALI induction in mice, we developed a physiologically based pharmacokinetic model for albumin nanoparticles (ANP). The model was calibrated using data from mice treated with intraperitoneal LPS (6 mg/kg), followed by intravenous ANP (0.5 mg/mouse or about 20.8 mg/kg) at 0.5, 6, and 24 h. The simulation results reproduced the experimental observations and indicated that the accumulation of ANP in the lungs increased, reaching a peak 6 h after LPS injury, whereas it decreased in the liver, kidney, and spleen. The model predicted that LPS caused an immediate (within the first 30 min) dramatic increase in lung and kidney tissue permeability, whereas splenic tissue permeability gradually increased over 24 h after LPS injection. This information can be used to design new therapies targeting specific organs affected by bacterial infections and potentially by other inflammatory insults.
Collapse
Affiliation(s)
- Elena O. Kutumova
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, 630090, Novosibirsk, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
| | - Ilya R. Akberdin
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090, Novosibirsk, Russia
| | - Vera S. Egorova
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Ekaterina P. Kolesova
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Alessandro Parodi
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Vadim S. Pokrovsky
- N.N. Blokhin Medical Research Center of Oncology, 115522, Moscow, Russia
- Patrice Lumumba People's Friendship University, 117198, Moscow, Russia
| | - Andrey A. Zamyatnin, Jr
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Faculty of Bioengineering and Bioinformatics and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234, Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Fedor A. Kolpakov
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, 630090, Novosibirsk, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
| |
Collapse
|
4
|
Liu D, Guo R, Shi B, Chen M, Weng S, Weng J. Fortunellin ameliorates LPS-induced acute lung injury, inflammation, and collagen deposition by restraining the TLR4/NF-κB/NLRP3 pathway. Immun Inflamm Dis 2024; 12:e1164. [PMID: 38501503 PMCID: PMC10949398 DOI: 10.1002/iid3.1164] [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: 05/17/2023] [Revised: 12/29/2023] [Accepted: 01/10/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE Acute lung injury (ALI) is the prevalent respiratory disease of acute inflammation with high morbidity and mortality. Fortunellin has anti-inflammation property, but its role in ALI remains elusive. Thus, this study clarified the function of fortunellin on ALI pathogenesis. METHODS The ALI mouse model was established by lipopolysaccharide (LPS) induction, and lung tissue damage was evaluated utilizing hematoxylin-eosin (HE) staining. The edema of lung tissue was measured by the lung wet/dry (W/D) ratio. The lung capillary permeability was reflected by the protein content in bronchoalveolar lavage fluid (BALF). Inflammatory cell infiltration was measured by the evaluation of the content of myeloperoxidase (MPO), neutrophils, and leukocytes in BALF. Cell apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The secretions of inflammatory cytokines were quantified using enzyme-linked immunosorbent assay (ELISA) assays. Lung tissue collagen deposition was evaluated by Masson staining. RESULTS Fortunellin attenuated LPS-induced lung tissue damage and reduced the W/D ratio, the content of MPO in lung tissue, the total protein contents in BALF, and the neutrophils and leukocytes number. Besides, fortunellin alleviated LPS-stimulated lung tissue apoptosis, inflammatory response, and collagen deposition. Furthermore, Fortunellin repressed the activity of the Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB)/NLR Family Pyrin Domain Containing 3 (NLRP3) pathway in the LPS-stimulated ALI model and LPS-induced RAW264.7 cells. Moreover, fortunellin attenuated LPS-stimulated tissue injury, apoptosis, inflammation, and collagen deposition of the lung via restraining the TLR4/NF-κB/NLRP3 pathway. CONCLUSION Fortunellin attenuated LPS-stimulated ALI through repressing the TLR4/NF-κB/NLRP3 pathway. Fortunellin may be a valuable drug for ALI therapy.
Collapse
Affiliation(s)
- Danjuan Liu
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Rongjie Guo
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Bingbing Shi
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Min Chen
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| | - Shuoyun Weng
- School of Ophthalmology & OptometryWenzhou Medical UniversityWenzhouChina
| | - Junting Weng
- Department of Critical Care Medicinethe Affiliated Hospital of Putian UniversityPutianChina
| |
Collapse
|
5
|
Jiaying Y, Bo S, Xiaolu W, Yanyan Z, Hongjie W, Nan S, Bo G, Linna W, Yan Z, Wenya G, Keke L, Shan J, Chuan L, Yu Z, Qinghe Z, Haiyu Z. Arenobufagin-loaded PEG-PLA nanoparticles for reducing toxicity and enhancing cancer therapy. Drug Deliv 2023; 30:2177362. [PMID: 36772846 PMCID: PMC9930844 DOI: 10.1080/10717544.2023.2177362] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Arenobufagin (ArBu) is a natural anticancer drug with good anti-tumor effects, but its clinical applications and drug development potential are limited due to its toxicity. The purpose of this study is to reduce the toxic side effects of ArBu and improve the efficacy of tumor treatment by incorporating it into poly(ethylene glycol)-b-poly (lactide) co-polymer (PEG-PLA). ArBu@PEG-PLA micelles were prepared by a thin film hydration method. The optimized micelles were characterized by size, stability, drug loading, encapsulation rate, and drug release. The tumor-inhibition efficacy of the micelles was evaluated on A549 cells and tumor-bearing mice. The ArBu@PEG-PLA micelles have good drug-loading capacity, release performance, and stability. They can accumulate at the tumor site through the EPR effect. The micelles induce apoptosis through a mitochondrial apoptosis pathway. Compared with the free ArBu, the ArBu@PEG-PLA micelles had lower toxicity and higher safety in the acute toxicity evaluation experiment. The in vivo anti-tumor experiment with tumor-bearing mice showed that the tumor-inhibition rate of ArBu@PEG-PLA micelles was 72.9%, which was 1.28-fold higher than that of free ArBu (57.1%), thus showing a good tumor treatment effect. This study indicates that ArBu@PEG-PLA polymeric micelles can significantly improve the toxicity and therapeutic efficacy of ArBu. These can lead to a new therapeutic strategy to reduce the toxicity of ArBu and enhance tumor treatment.
Collapse
Affiliation(s)
- Yang Jiaying
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Sun Bo
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Wei Xiaolu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Zhou Yanyan
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Wang Hongjie
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Si Nan
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Gao Bo
- China Resources Sanjiu Modern Traditional Chinese Medicine Pharmaceutical Co., Ltd, Shenzhen, China
| | - Wang Linna
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Zhang Yan
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Gao Wenya
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Luo Keke
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Jiang Shan
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| | - Luo Chuan
- Anhui Huarun Jinchan Pharmaceutical Co., Ltd, Anhui, China
| | - Zhao Yu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China,CONTACT Zhao Yu
| | - Zhao Qinghe
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China,Zhao Qinghe
| | - Zhao Haiyu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China,Zhao Haiyu China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing, China
| |
Collapse
|
6
|
Yang Y, Zoulikha M, Xiao Q, Huang F, Jiang Q, Li X, Wu Z, He W. Pulmonary endothelium-targeted nanoassembly of indomethacin and superoxide dismutase relieves lung inflammation. Acta Pharm Sin B 2023; 13:4607-4620. [PMID: 37969734 PMCID: PMC10638505 DOI: 10.1016/j.apsb.2023.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 11/17/2023] Open
Abstract
Lung inflammation is an essential inducer of various diseases and is closely related to pulmonary-endothelium dysfunction. Herein, we propose a pulmonary endothelium-targeted codelivery system of anti-inflammatory indomethacin (IND) and antioxidant superoxide dismutase (SOD) by assembling the biopharmaceutical SOD onto the "vector" of rod-like pure IND crystals, followed by coating with anti-ICAM-1 antibody (Ab) for targeting endothelial cells. The codelivery system has a 237 nm diameter in length and extremely high drug loading of 39% IND and 2.3% SOD. Pharmacokinetics and biodistribution studies demonstrate the extended blood circulation and the strong pulmonary accumulation of the system after intravenous injection in the lipopolysaccharide (LPS)-induced inflammatory murine model. Particularly, the system allows a robust capacity to target pulmonary endothelium mostly due to the rod-shape and Ab coating effect. In vitro, the preparation shows the synergistic anti-inflammatory and antioxidant effects in LPS-activated endothelial cells. In vivo, the preparation exhibits superior pharmacodynamic efficacy revealed by significantly downregulating the inflammatory/oxidative stress markers, such as TNF-α, IL-6, COX-2, and reactive oxygen species (ROS), in the lungs. In conclusion, the codelivery system based on rod-like pure crystals could well target the pulmonary endothelium and effectively alleviate lung inflammation. The study offers a promising approach to combat pulmonary endothelium-associated diseases.
Collapse
Affiliation(s)
- Yi Yang
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Makhloufi Zoulikha
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feifei Huang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qi Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| |
Collapse
|
7
|
Huang QF, Li YH, Huang ZJ, Jun M, Wang W, Chen XL, Wang GH. Artesunate carriers induced ferroptosis to overcome biological barriers for anti-cancer. Eur J Pharm Biopharm 2023; 190:284-293. [PMID: 37532638 DOI: 10.1016/j.ejpb.2023.07.014] [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: 06/20/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
Artesunate (ART) has potent anticancer activity but it suffers from poor stability and low bioavailability in vivo due to the special endoperoxide moiety in the molecules. In this work, we fabricated programmable enzyme/reactive oxygen species (ROS) responsive ART complex carriers with size and charge adaptive regulation in order to improve stability and overcome biochemical hurdles of solid tumor. The complex carries (ART/AA-PAMAM@HA) were created by electrostatic interaction between dendrimer-ART/arachidonic acid (AA) (ART/AA-PAMAM) and hyaluronic acid (HA), which can proactively penetrate deeply into tumors and selective drug release. Specifically, ART induced Fenton reaction and produced a mass of ROS and lipid peroxides (LPO), leading to the depressing of GSH level and glutathione peroxidase 4 (GPX4) activity. Meanwhile, exogenous AA further promoted the accumulation of LPO by cascade regulating ferroptosis pathway. In the anti-tumor efficacy in vivo, the tumor inhibition ratio was achieved to 46.92%. This work shows a new anti-tumor strategy triggering ferroptosis via regulating redox homeostasis.
Collapse
Affiliation(s)
- Qun-Fa Huang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, China; School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yan-Hong Li
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, China; School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zeng-Jin Huang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, China; School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Mei Jun
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wei Wang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Xiao-Li Chen
- Guangzhou Institute of Energy Conversion, Chinese Academy of Sciences, Guangzhou 510640, Guangdong, China
| | - Guan-Hai Wang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, 523710, Dongguan, China; School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
8
|
Holloman BL, Cannon A, Wilson K, Nagarkatti P, Nagarkatti M. Aryl Hydrocarbon Receptor Activation Ameliorates Acute Respiratory Distress Syndrome through Regulation of Th17 and Th22 Cells in the Lungs. mBio 2023; 14:e0313722. [PMID: 36809070 PMCID: PMC10128024 DOI: 10.1128/mbio.03137-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 02/23/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is triggered by a variety of insults, including bacterial and viral infections, and this leads to high mortality. While the role of the aryl hydrocarbon receptor (AhR) in mucosal immunity is being increasingly recognized, its function during ARDS is unclear. In the current study, we investigated the role of AhR in LPS-induced ARDS. AhR ligand, indole-3-carbinol (I3C), attenuated ARDS which was associated with a decrease in CD4+ RORγt +IL-17a+IL-22+ pathogenic Th17 cells, but not CD4+RORγt +IL-17a+IL-22- homeostatic Th 17 cells, in the lungs. AhR activation also led to a significant increase in CD4+IL-17a-IL-22+ Th22 cells. I3C-mediated Th22 cell expansion was dependent on the AhR expression on RORγt+ cells. AhR activation downregulated miR-29b-2-5p in immune cells from the lungs, which in turn downregulated RORc expression and upregulated IL-22. Collectively, the current study suggests that AhR activation can attenuate ARDS and may serve as a therapeutic modality by which to treat this complex disorder. IMPORTANCE Acute respiratory distress syndrome (ARDS) is a type of respiratory failure that is triggered by a variety of bacterial and viral infections, including the coronavirus SARS-CoV2. ARDS is associated with a hyperimmune response in the lungs that which is challenging to treat. Because of this difficulty, approximately 40% of patients with ARDS die. Thus, it is critical to understand the nature of the immune response that is functional in the lungs during ARDS as well as approaches by which to attenuate it. AhR is a transcription factor that is activated by a variety of endogenous and exogenous environmental chemicals as well as bacterial metabolites. While AhR has been shown to regulate inflammation, its role in ARDS is unclear. In the current study, we provide evidence that AhR activation can attenuate LPS-mediated ARDS through the activation of Th22 cells in the lungs, which are regulated through miR-29b-2-5p. Thus, AhR can be targeted to attenuate ARDS.
Collapse
Affiliation(s)
- Bryan Latrell Holloman
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Alkeiver Cannon
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Kiesha Wilson
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
9
|
Casagrande LDR, Porto GD, Colares MC, Venturini LM, Silveira GDB, Mendes C, Corrêa MEAB, Lima IR, Feuser PE, Machado-de-Ávila RA, Silveira PCL. Green synthesis of gold nanoparticles modulates lipopolysaccharide-induced lung inflammation in Wistar rats. Basic Clin Pharmacol Toxicol 2023; 132:473-485. [PMID: 36882317 DOI: 10.1111/bcpt.13854] [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: 01/12/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023]
Abstract
This study aimed to investigate the effect of intranasal treatment of gold nanoparticles (GNPs) and Curcumin (Cur) on the lipopolysaccharide (LPS)-induced acute pulmonary inflammatory response. A single intraperitoneal injection of LPS (0.5 mg/Kg) was performed, and the animals in the Sham group were injected with 0.9% saline. Treatment was daily intranasally with GNPs (2.5 mg/L), Cur (10 mg/kg) and GNP-Cur started 12 h after LPS administration and ended on the seventh day. The results show that the treatment performed with GNP-Cur was the most effective to attenuate the action of pro-inflammatory cytokines, and a lower leukocyte count in the bronchoalveolar lavage, in addition to positively regulating anti-inflammatory cytokines in relation to other groups. As a result, it promoted an oxirreductive balanced environment in the lung tissue, providing a histological outcome with a reduction in inflammatory cells and greater alveolar area. The group treated with GNPs-Cur was superior to the other groups, with better anti-inflammatory activity and reduced oxidative stress, resulting in less morphological damage to lung tissue. In conclusion, the use of reduced GNPs with curcumin demonstrates promising effects in the control of the acute inflammatory response, helping to protect the lung tissue at the biochemical and morphological levels.
Collapse
Affiliation(s)
- Laura de R Casagrande
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Germano D Porto
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Mateus C Colares
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Ligia M Venturini
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Gustavo de B Silveira
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Carolini Mendes
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Maria Eduarda A B Corrêa
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Igor R Lima
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Paulo E Feuser
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Ricardo A Machado-de-Ávila
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| | - Paulo C L Silveira
- Laboratory of Experimental Physiopathology, Program of postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma, Santa Catarina, 88806-000, Brazil
| |
Collapse
|
10
|
Deng L, Jian Z, Xu T, Li F, Deng H, Zhou Y, Lai S, Xu Z, Zhu L. Macrophage Polarization: An Important Candidate Regulator for Lung Diseases. Molecules 2023; 28:molecules28052379. [PMID: 36903624 PMCID: PMC10005642 DOI: 10.3390/molecules28052379] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Macrophages are crucial components of the immune system and play a critical role in the initial defense against pathogens. They are highly heterogeneous and plastic and can be polarized into classically activated macrophages (M1) or selectively activated macrophages (M2) in response to local microenvironments. Macrophage polarization involves the regulation of multiple signaling pathways and transcription factors. Here, we focused on the origin of macrophages, the phenotype and polarization of macrophages, as well as the signaling pathways associated with macrophage polarization. We also highlighted the role of macrophage polarization in lung diseases. We intend to enhance the understanding of the functions and immunomodulatory features of macrophages. Based on our review, we believe that targeting macrophage phenotypes is a viable and promising strategy for treating lung diseases.
Collapse
Affiliation(s)
- Lishuang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
| | - Zhijie Jian
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
| | - Tong Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
| | - Fengqin Li
- College of Animal Science, Xichang University, Xichang 615000, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
| | - Yuancheng Zhou
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 625014, China
| | - Siyuan Lai
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 625014, China
- Correspondence: (Z.X.); (L.Z.); Tel.: +86-139-8160-4765 (L.Z.)
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 625014, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 625014, China
- Correspondence: (Z.X.); (L.Z.); Tel.: +86-139-8160-4765 (L.Z.)
| |
Collapse
|