1
|
Paris-Robidas S, Bolduc I, Lapointe V, Galimi J, Lemieux P, Huppé CA, Couture F. Impact of time intervals on drug efficacy and phenotypic outcomes in acute respiratory distress syndrome in mice. Sci Rep 2024; 14:20768. [PMID: 39237657 PMCID: PMC11377577 DOI: 10.1038/s41598-024-71659-x] [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: 04/12/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024] Open
Abstract
Acute respiratory distress syndrome is a severe lung condition resulting from various causes, with life-threatening consequences that necessitate intensive care. The phenomenon can be modeled in preclinical models, notably through the use of lipopolysaccharide (LPS) instillation in mice. The phenotype induced closely recapitulates the human syndrome, including pulmonary edema, leukocyte infiltration, acute inflammation, impaired pulmonary function, and histological damage. However, the experimental designs using LPS instillations are extremely diverse in the literature. This highly complicates the interpretation of the induced phenotype chronology for future study design and hinders the proper identification of the optimal time frame to assess different readouts. Therefore, the definition of the treatment window in relation to the beginning of the disease onset also presents a significant challenge to address questions or test compound efficacy. In this context, the temporality of the different readouts usually measured in the model was evaluated in both normal and neutrophil-depleted male C57bl/6 mice using LPS-induction to assess the best window for proper readout evaluation with an optimal dynamic response range. Ventilation parameters were evaluated by whole-body plethysmography and neutrophil recruitment were evaluated in bronchoalveolar lavage fluids and in lung tissues directly. Imaging evaluation of myeloperoxidase along with activity in lung lysates and fluids were compared, along with inflammatory cytokines and lung extravasation by enzyme-linked immunoassays. Moreover, dexamethasone, the gold standard positive control in this model, was also administered at different times before and after phenotype induction to assess how kinetics affected each parameter. Overall, our data demonstrate that each readout evaluated in this study has a singular kinetic and highlights the key importance of the timing between ARDS phenotype and treatment administration and/or analysis. These findings also strongly suggest that analyzes, both in-life and post-mortem should be conducted at multiple time points to properly capture the dynamic phenotype of the LPS-ARDS model and response to treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Frédéric Couture
- TransBIOTech, Lévis, QC, G6V 6Z3, Canada.
- Nutraceuticals and Functional Foods Institute (INAF), Université Laval, Québec City, QC, G1K 7P4, Canada.
- Centre Intégré de Santé Et de Services Sociaux de Chaudière-Appalaches, Lévis, QC, G6E 3E2, Canada.
| |
Collapse
|
2
|
Lopuszynski J, Wang J, Zahid M. Beyond Transduction: Anti-Inflammatory Effects of Cell Penetrating Peptides. Molecules 2024; 29:4088. [PMID: 39274936 PMCID: PMC11397606 DOI: 10.3390/molecules29174088] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
One of the bottlenecks to bringing new therapies to the clinic has been a lack of vectors for delivering novel therapeutics in a targeted manner. Cell penetrating peptides (CPPs) have received a lot of attention and have been the subject of numerous developments since their identification nearly three decades ago. Known for their transduction abilities, they have generally been considered inert vectors. In this review, we present a schema for their classification, highlight what is known about their mechanism of transduction, and outline the existing literature as well as our own experience, vis a vis the intrinsic anti-inflammatory properties that certain CPPs exhibit. Given the inflammatory responses associated with viral vectors, CPPs represent a viable alternative to such vectors; furthermore, the anti-inflammatory properties of CPPs, mostly through inhibition of the NF-κB pathway, are encouraging. Much more work in relevant animal models, toxicity studies in large animal models, and ultimately human trials are needed before their potential is fully realized.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Department of Cardiovascular Medicine, Guggenheim Gu 9-01B, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Dilxat T, Shi Q, Chen X, Liu X. Garlic oil supplementation blocks inflammatory pyroptosis-related acute lung injury by suppressing the NF-κB/NLRP3 signaling pathway via H 2S generation. Aging (Albany NY) 2024; 16:6521-6536. [PMID: 38613798 PMCID: PMC11042940 DOI: 10.18632/aging.205721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/09/2024] [Indexed: 04/15/2024]
Abstract
Acute lung injury (ALI) is a major cause of acute respiratory failure with a high morbidity and mortality rate, and effective therapeutic strategies for ALI remain limited. Inflammatory response is considered crucial for the pathogenesis of ALI. Garlic, a globally used cooking spice, reportedly exhibits excellent anti-inflammatory bioactivity. However, protective effects of garlic against ALI have never been reported. This study aimed to investigate the protective effects of garlic oil (GO) supplementation on lipopolysaccharide (LPS)-induced ALI models. Hematoxylin and eosin staining, pathology scores, lung myeloperoxidase (MPO) activity measurement, lung wet/dry (W/D) ratio detection, and bronchoalveolar lavage fluid (BALF) analysis were performed to investigate ALI histopathology. Real-time polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to evaluate the expression levels of inflammatory factors, nuclear factor-κB (NF-κB), NLRP3, pyroptosis-related proteins, and H2S-producing enzymes. GO attenuated LPS-induced pulmonary pathological changes, lung W/D ratio, MPO activity, and inflammatory cytokines in the lungs and BALF. Additionally, GO suppressed LPS-induced NF-κB activation, NLRP3 inflammasome expression, and inflammatory-related pyroptosis. Mechanistically, GO promoted increased H2S production in lung tissues by enhancing the conversion of GO-rich polysulfide compounds or by increasing the expression of H2S-producing enzymes in vivo. Inhibition of endogenous or exogenous H2S production reversed the protective effects of GO on ALI and eliminated the inhibitory effects of GO on NF-κB, NLRP3, and pyroptotic signaling pathways. Overall, these findings indicate that GO has a critical anti-inflammatory effect and protects against LPS-induced ALI by suppressing the NF-κB/NLRP3 signaling pathway via H2S generation.
Collapse
Affiliation(s)
- Tursunay Dilxat
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Qiang Shi
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Xiaofan Chen
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Xuxin Liu
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| |
Collapse
|
4
|
Nguyen TT, Deng Z, Guo RY, Chai JW, Li R, Zeng QY, Lai SA, Chen X, Xu XQ. Periplaneta Americana Extract Ameliorates LPS-induced Acute Lung Injury Via Reducing Inflammation and Oxidative Stress. Curr Med Sci 2023:10.1007/s11596-023-2723-8. [PMID: 37191939 DOI: 10.1007/s11596-023-2723-8] [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: 05/05/2022] [Accepted: 12/23/2022] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Acute lung injury (ALI) is an acute clinical syndrome characterized by uncontrolled inflammation response, which causes high mortality and poor prognosis. The present study determined the protective effect and underlying mechanism of Periplaneta americana extract (PAE) against lipopolysaccharide (LPS)-induced ALI. METHODS The viability of MH-S cells was measured by MTT. ALI was induced in BALB/c mice by intranasal administration of LPS (5 mg/kg), and the pathological changes, oxidative stress, myeloperoxidase activity, lactate dehydrogenase activity, inflammatory cytokine expression, edema formation, and signal pathway activation in lung tissues and bronchoalveolar lavage fluid (BALF) were examined by H&E staining, MDA, SOD and CAT assays, MPO assay, ELISA, wet/dry analysis, immunofluorescence staining and Western blotting, respectively. RESULTS The results revealed that PAE obviously inhibited the release of proinflammatory TNF-α, IL-6 and IL-1β by suppressing the activation of MAPK/Akt/NF-κB signaling pathways in LPS-treated MH-S cells. Furthermore, PAE suppressed the neutrophil infiltration, permeability increase, pathological changes, cellular damage and death, pro-inflammatory cytokines expression, and oxidative stress upregulation, which was associated with its blockage of the MAPK/Akt/NF-κB pathway in lung tissues of ALI mice. CONCLUSION PAE may serve as a potential agent for ALI treatment due to its anti-inflammatory and anti-oxidative properties, which correlate to the blockage of the MAPK/NF-κB and AKT signaling pathways.
Collapse
Affiliation(s)
- Tien-Thanh Nguyen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ze Deng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Rui-Yin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jin-Wei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qing-Ye Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shi-An Lai
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Xue-Qing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Kim Y, Bae CR, Kim D, Kim H, Lee S, Zhang H, Noh M, Kim YM, Mochizuki N, Kwon YG. Efficacy of CU06-1004 via regulation of inflammation and endothelial permeability in LPS-induced acute lung injury. J Inflamm (Lond) 2023; 20:13. [PMID: 37024954 PMCID: PMC10078077 DOI: 10.1186/s12950-023-00338-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a life-threatening condition that fundamentally results from inflammation and edema in the lung. There are no effective treatments available for clinical use. Previously, we found that as a leakage blocker CU06-1004 prevents endothelial barrier disruption and enhances endothelial cell survival under inflammatory conditions. In this study, we aimed to elucidate the effect of CU06-1004 in terms of prevention of inflammation and endothelial dysfunction in an ALI mouse model. METHODS An ALI model was established that included intraperitoneal administration of LPS. Following LPS administration, survival rates and lung wet/dry ratios were assessed. Histological analysis was performed using hematoxylin and eosin staining. Scanning electron microscopy was used to examine alveolar and capillary morphology. Cytokines such as IL-1β, IL-6, and TNF-α were analyzed using an ELISA assay of bronchoalveolar lavage fluid (BALF) and serum. Neutrophil infiltration was observed in BALF using Wright-Giemsa staining, and myeloperoxidase (MPO) activity was assessed. Pulmonary vascular leakage was confirmed using Evans-blue dye, and the expression of junctional proteins was evaluated using immunofluorescent staining. Expression of adhesion molecules was observed using immunofluorescence staining. NF-κB activation was determined using immunohistochemistry and western blot analysis. RESULTS Survival rates and pulmonary edema were ameliorated with CU06-1004 treatment. Administration of CU06-1004 normalized histopathological changes induced by LPS, and alveolar-capillary wall thickening was reduced. Compared with the LPS-challenged group, after CU06-1004 treatment, the infiltration of immune cells was decreased in the BALF, and MPO activity in lung tissue was reduced. Similarly, in the CU06-1004 treatment group, pro-inflammatory cytokines were significantly inhibited in both BALF and serum. Evans-blue leakage was reduced, and the expression of junctional proteins was recovered in the CU06-1004 group. Adhesion molecules were downregulated and NF-κB activation was inhibited after CU06-1004 treatment. CONCLUSIONS These results suggested that CU06-1004 had a therapeutic effect against LPS-induced ALI via alleviation of the inflammatory response and protection of vascular integrity.
Collapse
Affiliation(s)
- Yeomyeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Cho-Rong Bae
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Dongyeop Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyejeong Kim
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Sunghye Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Haiying Zhang
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Minyoung Noh
- Department of Bio Research, Curacle Co. Ltd, Seoul, 06694, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe- shimmachi, Suita, Osaka, 564-8565, Japan
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
6
|
Wu Z, Chen L, Wang Q, Govindasamy C, Subramaniyan Sivakumar A, Chen X. Betanin Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Mice via Inhibition of Inflammatory Response and Oxidative Stress. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
7
|
Li Y, Wang SM, Li X, Lv CJ, Peng LY, Yu XF, Song YJ, Wang CJ. Pterostilbene pre-treatment reduces LPS-induced acute lung injury through activating NR4A1. PHARMACEUTICAL BIOLOGY 2022; 60:394-403. [PMID: 35271397 PMCID: PMC8920364 DOI: 10.1080/13880209.2022.2034893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 01/05/2022] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT Pterostilbene (PTE), a common polyphenol compound, exerts an anti-inflammatory effect in many diseases, including acute lung injury (ALI). OBJECTIVE This study explores the potential mechanism of PTE pre-treatment against lipopolysaccharide (LPS)-induced ALI. MATERIALS AND METHODS Sixty Sprague-Dawley rats were divided into control, ALI, 10 mg/kg PTE + LPS, 20 mg/kg PTE + LPS, and 40 mg/kg PTE + LPS groups. At 24 h before LPS instillation, PTE was administered orally. At 2 h before LPS instillation, PTE was again administered orally. After 24 h of LPS treatment, the rats were euthanized. The levels of inflammatory cells and inflammatory factors in the bronchoalveolar lavage fluid (BALF), the expression of nuclear receptor subfamily 4 group A member 1 (NR4A1), and the nuclear factor (NF)-κB pathway-related protein levels were detected. NR4A1 agonist was used to further investigate the mechanism of PTE pre-treatment. RESULTS After PTE pre-treatment, the LPS induced inflammation was controlled and the survival rate was increased to 100% from 70% after LPS treatment 24 h. For lung injury score, it decreased to 1.5 from 3.5 after treating 40 mg/kg PTE. Compared with the control group, the expression of NR4A1 in the ALI group was decreased by 20-40%. However, the 40 mg/kg PTE pre-treatment increased the NR4A1 expression by 20-40% in the lung tissue. The results obtained with pre-treatment NR4A1 agonist were similar to those obtained by pre-treatment 40 mg/kg PTE. CONCLUSIONS PTE pre-treatment might represent an appropriate therapeutic target and strategy for preventing ALI induced by LPS.
Collapse
Affiliation(s)
- Ying Li
- Department of Emergency, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Shu-Min Wang
- Department of Emergency, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Xing Li
- Department of Station Intergrate Service, Yantai Central Blood, Yantai, Shandong, China
| | - Chang-Jun Lv
- Binzhou Medical University, Yantai, Shandong, China
| | - Ling-Yun Peng
- Department of Thoracic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Xiao-Feng Yu
- Department of Thoracic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Ying-Jian Song
- Department of Thoracic Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Cong-Jie Wang
- Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
8
|
Mo Y, Kang SY, Bang JY, Kim Y, Jeong J, Jeong EM, Kim HY, Cho SH, Kang HR. Intravenous Mesenchymal Stem Cell Administration Modulates Monocytes/Macrophages and Ameliorates Asthmatic Airway Inflammation in a Murine Asthma Model. Mol Cells 2022; 45:833-845. [PMID: 36380733 PMCID: PMC9676992 DOI: 10.14348/molcells.2022.0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Although asthma is a common chronic airway disease that responds well to anti-inflammatory agents, some patients with asthma are unresponsive to conventional treatment. Mesenchymal stem cells (MSCs) have therapeutic potential for the treatment of inflammatory diseases owing to their immunomodulatory properties. However, the target cells of MSCs are not yet clearly known. This study aimed to determine the effect of human umbilical cord-derived MSCs (hUC-MSCs) on asthmatic lungs by modulating innate immune cells and effector T cells using a murine asthmatic model. Intravenously administered hUC-MSCs reduced airway resistance, mucus production, and inflammation in the murine asthma model. hUC-MSCs attenuated not only T helper (Th) 2 cells and Th17 cells but also augmented regulatory T cells (Tregs). As for innate lymphoid cells (ILC), hUC-MSCs effectively suppressed ILC2s by downregulating master regulators of ILC2s, such as Gata3 and Tcf7. Finally, regarding lung macrophages, hUC-MSCs reduced the total number of macrophages, particularly the proportion of the enhanced monocyte-derived macrophage population. In a closer examination of monocyte-derived macrophages, hUC-MSCs reduced the M2a and M2c populations. In conclusion, hUC-MSCs can be considered as a potential anti- asthmatic treatment given their therapeutic effect on the asthmatic airway inflammation in a murine asthma model by modulating innate immune cells, such as ILC2s, M2a, and M2c macrophages, as well as affecting Tregs and effector T cells.
Collapse
Affiliation(s)
- Yosep Mo
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung-Yoon Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Korea
| | - Ji-Young Bang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yujin Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jiung Jeong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eui-Man Jeong
- Department of Pharmacy, Jeju National University College of Pharmacy, Jeju 63243, Korea
| | - Hye Young Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Medical Science, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang-Heon Cho
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
9
|
Mishra P, Pandey R, Pandey N, Tripathi S, Tripathi YB. Prevention of mortality in acute lung injury induced by oleic acid: Application of polyherbal decoction (bronco T). Front Cell Dev Biol 2022; 10:1003767. [PMID: 36313556 PMCID: PMC9612945 DOI: 10.3389/fcell.2022.1003767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Acute lung injury (ALI) is a lethal respiratory disorder; directed uncontrolled inflammation, sloughing of the alveolar cells and their diffusion, and altered cardiorespiratory parameters with a global mortality rate of 40%. This study was designed to assess the preventive effect of a polyherbal decoction (Bronco T, 1.5 g/kg b. w.) on cardiorespiratory variables in oleic acid-induced ALI in rats. Oleic acid increases the level of neutrophil infiltration leading to pulmonary edema and alters the cardiorespiratory dynamics. The adult male rats were surgically cannulated and treated with intravenous oleic acid (0.38 ml/kg b. w.) to establish the ALI model. Bronco T was pre-administered orally 3 hours before oleic acid. The biophysical, histological, biochemical, and molecular effects were compared with dexamethasone (5 mg/kg b. w. i. p.). The animals were randomly divided into control, lethal, standard, and treatment groups. Respiratory frequency (RF), heart rate (HR), and mean arterial pressure (MAP) were recorded on a computerized chart recorder; arterial blood sample was collected to determine PaO2/FiO2, TNF-α, and MPO. Lipid peroxidation, superoxide dismutase, and catalase activity were evaluated to measure oxidative stress in bronchoalveolar lavage. Additionally, the pulmonary water content, COX-2 expression and histological examination were determined in the lung. A molecular docking study of the active phytoconstituent of BT obtained from HR-LCMS analysis against reported targets (IL-6, COX-2, TNFα, MPO and ENaC) of ALI was carried out. The B.T. pretreatment prevents mortality in comparison to the oleic acid group. It protects the lungs and heart from the detrimental effect of oleic acid, on par with dexamethasone. COX-2 mRNA expression was significantly down-regulated in the treatment group. The reduced level of TNF-α, MPO, SOD and catalase supported the protective effect of B.T. The in silico study revealed strong binding interaction between the phytoconstituent (Galangin 3- [galactosyl-(1–4)-rhamnoside and Beta solamarine] of BT and the reported target. The B.T. pre-administration attenuates the oleic acid-induced mortality and cardiorespiratory toxicity.
Collapse
Affiliation(s)
- Priyanka Mishra
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ratna Pandey
- Department of Physiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Nikhil Pandey
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Suyash Tripathi
- Department of Cardiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Yamini Bhusan Tripathi
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Yamini Bhusan Tripathi,
| |
Collapse
|
10
|
Zhang J, Chang J, Beg MA, Huang W, Zhao Y, Dai W, Wu X, Cui W, Pillai SS, Lakhani HV, Sodhi K, Shapiro JI, Sahoo D, Zheng Z, Silverstein RL, Chen Y. Na/K-ATPase suppresses LPS-induced pro-inflammatory signaling through Lyn. iScience 2022; 25:104963. [PMID: 36072548 PMCID: PMC9442361 DOI: 10.1016/j.isci.2022.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/02/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Na/K-ATPase (NKA), besides its ion transporter function, is a signal transducer by regulating Src family kinases (SFK). The signaling NKA contributes to oxidized LDL-induced macrophage foam cell formation and interacts with TLR4. However, its role in lipopolysaccharides (LPS)-induced signaling and glycolytic switch in macrophages remains unclear. Using peritoneal macrophages from NKA α1 haploinsufficient mice (NKA α1+/-), we found that NKA α1 haploinsufficiency led to enhanced LPS-stimulated NF-κB pathway, ROS signaling, and pro-inflammatory cytokines. Intraperitoneal injection of LPS resulted in more severe lung inflammation and injury with lower survival rate in NKA α1+/- mice. Additionally, LPS induced a higher extent of the metabolic switch from oxidative phosphorylation to glycolysis. Mechanistically, NKA α1 interacted with TLR4 and Lyn. The presence of NKA α1 in this complex attenuated Lyn activation by LPS, which subsequently restricted the downstream ROS and NF-κB signaling. In conclusion, we demonstrated that NKA α1 suppresses LPS-induced macrophage pro-inflammatory signaling through Lyn.
Collapse
Affiliation(s)
- Jue Zhang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Jackie Chang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | | | - Wenxin Huang
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Yiqiong Zhao
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Wen Dai
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Xiaopeng Wu
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| | - Weiguo Cui
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sneha S. Pillai
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Hari Vishal Lakhani
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Komal Sodhi
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Daisy Sahoo
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ze Zheng
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Roy L. Silverstein
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
11
|
Peng K, Deng N, Meng Y, He Q, Meng H, Luo T, Wei Y, Kang Y, Zhou X, Shen F. Alpha-Momorcharin Inhibits Proinflammatory Cytokine Expression by M1 Macrophages but Not Anti-Inflammatory Cytokine Expression by M2 Macrophages. J Inflamm Res 2022; 15:4853-4872. [PMID: 36042868 PMCID: PMC9420447 DOI: 10.2147/jir.s372306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Alpha-momorcharin (α-MMC) is a natural medicine derived from bitter melon and has been found to exert immunomodulatory effects. Our previous study indicated that α-MMC can regulate cytokine release from monocytes, but it remains unknown about its regulatory effect on different types of cytokines, such as inflammatory cytokines or anti-inflammatory cytokines. Methods LPS-induced M1-type macrophages model and IL-4-induced M2-type macrophages model were established, and the expression of proinflammatory cytokines and anti-inflammatory cytokines were assessed by ELISA after α-MMC was administered. Then, a LPS-induced acute pneumonia mouse model was established, the proinflammatory cytokines levels and inflammatory lesions in lung tissues were examined by ELISA or H&E staining. Furthermore, omics screening analysis and Western blotting verification were performed on TLR4 and JAK1-STAT6 signalling pathway-related proteins to elucidate the regulatory mechanism of α-MMC in those M1 macrophages and M2 macrophages. Results At a noncytotoxic dose of 0.3 μg/mL, α-MMC significantly inhibited the LPS-induced expression of inflammatory cytokines, such as TNF-α, IL-1β, IL-6, IL-8, MIP-1α and MCP-1, by M1 macrophages in a time-dependent manner, but α-MMC did not inhibit the IL-4-induced synthesis of anti-inflammatory cytokines, such as IL-10, IL-1RA, EGF, VEGF, TGF-β and CCL22, by M2 macrophages. Moreover, α-MMC also inhibited inflammatory cytokine expression in an LPS-induced acute pneumonia mouse model and alleviated inflammation in lung tissues. Furthermore, omics screening and Western blotting analysis confirmed that α-MMC inhibited TAK1/p-TAK1 and subsequently blocked the downstream MAPK and NF-κB pathways, thus inhibiting the LPS-induced inflammatory cytokine expression. Conclusion Our results reveal that α-MMC inhibits proinflammatory cytokine expression by M1 macrophages but not anti-inflammatory cytokine expression by M2 macrophages. The efficacy of α-MMC in selectively inhibiting proinflammatory cytokine expression renders it particularly suitable for the treatment of severe inflammation and autoimmune diseases characterized by cytokine storms.
Collapse
Affiliation(s)
- Kejun Peng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Nianhua Deng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Yao Meng
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Qianchuan He
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Hao Meng
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University School of Life Sciences, Beijing, People's Republic of China
| | - Ting Luo
- Wuhan Corebiolab Co., Ltd, Wuhan, People's Republic of China
| | - Yanru Wei
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Yue Kang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Xiaodong Zhou
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| | - Fubing Shen
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, People's Republic of China
| |
Collapse
|
12
|
Tao Q, Zhang ZD, Qin Z, Liu XW, Li SH, Bai LX, Ge WB, Li JY, Yang YJ. Aspirin eugenol ester alleviates lipopolysaccharide-induced acute lung injury in rats while stabilizing serum metabolites levels. Front Immunol 2022; 13:939106. [PMID: 35967416 PMCID: PMC9372404 DOI: 10.3389/fimmu.2022.939106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aspirin eugenol ester (AEE) was a novel drug compound with aspirin and eugenol esterified. AEE had various pharmacological activities, such as anti-inflammatory, antipyretic, analgesic, anti-oxidative stress and so on. In this study, it was aimed to investigate the effect of AEE on the acute lung injury (ALI) induced by lipopolysaccharide (LPS) in rats. In vitro experiments evaluated the protective effect of AEE on the LPS-induced A549 cells. The tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) were measured in the cell supernatant. The Wistar rats were randomly divided into five groups (n = 8): control group, model group (LPS group), LPS + AEE group (AEE, 54 mg·kg-1), LPS + AEE group (AEE, 108 mg·kg-1), LPS + AEE group (AEE, 216 mg·kg-1). The lung wet-to-dry weight (W/D) ratio and immune organ index were calculated. WBCs were counted in bronchoalveolar lavage fluid (BALF) and total protein concentration was measured. Hematoxylin-Eosin (HE) staining of lung tissue was performed. Glutathione (GSH), glutathione peroxidase (GPx), catalase (CAT), antioxidant superoxide dismutase (SOD), total antioxidant capacity (T-AOC), lactate dehydrogenase (LDH), C-reactive protein (CRP), myeloperoxidase (MPO), malondialdehyde (MDA), macrophage mobility inhibitory factor (MIF), TNF-α, IL-6, and IL-1β activity were measured. The metabolomic analysis of rat serum was performed by UPLC-QTOF-MS/MS. From the results, compared with LPS group, AEE improved histopathological changes, reduced MDA, CRP, MPO, MDA, and MIF production, decreased WBC count and total protein content in BALF, pro-inflammatory cytokine levels, immune organ index and lung wet-dry weight (W/D), increased antioxidant enzyme activity, in a dose-dependent manner. The results of serum metabolomic analysis showed that the LPS-induced ALI caused metabolic disorders and oxidative stress in rats, while AEE could ameliorate it to some extent. Therefore, AEE could alleviate LPS-induced ALI in rats by regulating abnormal inflammatory responses, slowing down oxidative stress, and modulating energy metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
13
|
Deuterated Arachidonic Acid Ameliorates Lipopolysaccharide-Induced Lung Damage in Mice. Antioxidants (Basel) 2022; 11:antiox11040681. [PMID: 35453366 PMCID: PMC9027010 DOI: 10.3390/antiox11040681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
Arachidonic acid (ARA) is a major component of lipid bilayers as well as the key substrate for the eicosanoid cascades. ARA is readily oxidized, and its non-enzymatic and enzymatic oxidation products induce inflammatory responses in nearly all tissues, including lung tissues. Deuteration at bis-allylic positions substantially decreases the overall rate of ARA oxidation when hydrogen abstraction is an initiating event. To compare the effects of dosing of arachidonic acid (H-ARA) and its bis-allylic hexadeuterated form (D-ARA) on lungs in conventionally healthy mice and in an acute lung injury model, mice were dosed with H-ARA or D-ARA for six weeks through dietary supplementation and then challenged with intranasal lipopolysaccharide (LPS) for subsequent analysis of bronchoalveolar lavage fluid and lung tissue. Dosing on D-ARA resulted in successful incorporation of D-ARA into various tissues. D-ARA significantly reduced LPS-induced adverse effects on alveolar septal thickness and the bronchoalveolar area. Oral deuterated ARA is taken up efficiently and protects against adverse LPS-induced pathology. This suggests novel therapeutic avenues for reducing lung damage during severe infections and other pathological conditions with inflammation in the pulmonary system and other inflammatory diseases.
Collapse
|
14
|
Mo Y, Kim Y, Bang JY, Jung J, Lee CG, Elias JA, Kang HR. Mesenchymal Stem Cells Attenuate Asthmatic Inflammation and Airway Remodeling by Modulating Macrophages/Monocytes in the IL-13-Overexpressing Mouse Model. Immune Netw 2022; 22:e40. [DOI: 10.4110/in.2022.22.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yosep Mo
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yujin Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Young Bang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jiung Jung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chun-Geun Lee
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Jack A. Elias
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Baicalin Liposome Alleviates Lipopolysaccharide-Induced Acute Lung Injury in Mice via Inhibiting TLR4/JNK/ERK/NF- κB Pathway. Mediators Inflamm 2020; 2020:8414062. [PMID: 33223957 PMCID: PMC7673921 DOI: 10.1155/2020/8414062] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/22/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are challenging diseases with the high mortality in a clinical setting. Baicalin (BA) is the main effective constituent isolated from the Chinese medical herb Scutellaria baicalensis Georgi, and studies have proved that it has a protective effect on ALI induced by lipopolysaccharide (LPS) due to the anti-inflammatory efficacy. However, BA has low solubility which may limit its clinical application. Hence, we prepared a novel drug delivery system—Baicalin liposome (BA-LP) in previous research—which can improve some physical properties of BA. Therefore, we aimed to explore the effect of BA-LP on ALI mice induced by LPS. In pharmacokinetics study, the values of t1/2 and AUC0-t in the BA-LP group were significantly higher than that of the BA group in normal mice, indicating that BA-LP could prolong the duration time in vivo of BA. The BA-LP group also showed a higher concentration in lung tissues than the BA group. Pharmacodynamics studies showed that BA-LP had a better effect than the BA group at the same dosage on reducing the W/D ratio, alleviating the lung injury score, and decreasing the proinflammatory factors (TNF-α, IL-1β) and total proteins in bronchoalveolar lavage fluids (BALF). In addition, the therapeutic effects of BA-LP showed a dose-dependent manner. Western blot analysis indicated that the anti-inflammatory action of BA could be attributed to the inhibition of the TLR4-NFκBp65 and JNK-ERK signaling pathways. These results suggest that BA-LP could be a valuable therapeutic candidate in the treatment of ALI.
Collapse
|
16
|
Toshchakov VY, Javmen A. Targeting the TLR signalosome with TIR domain-derived cell-permeable decoy peptides: the current state and perspectives. Innate Immun 2020; 26:35-47. [PMID: 31955621 PMCID: PMC6974878 DOI: 10.1177/1753425919844310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability to engineer pharmaceuticals that target the signal-dependent
interactions of signaling proteins should revolutionize drug development. One
approach to the rational design of protein interaction inhibitors uses decoy
peptides, i.e. segments of protein primary sequence, which are derived from
interfaces that mediate functional protein interactions. Decoy peptides often
retain the ability of the full-length prototype to bind the docking site of the
folded protein and thereby block the signal transduction. This review summarizes
advances made in the last decade in the development of cell-permeable decoy
peptide (CPDP) inhibitors to target the Toll/IL-1R resistance (TIR)
domain-mediated protein interactions in TLR signaling, in connection with the
recent progress in understanding of the TLR signalosome assembly mechanisms. We
present a large collection of currently available, TIR-targeting CPDPs and
propose their classification based on the types of TIR–TIR interactions they
target. The binding behavior of different CPDP-TIR pairs, studied in cell-based
assays and in binary in vitro systems using recombinant TIR
domains, is also reviewed. The available affinity data provide benchmarks for
rapid preliminary evaluation of future inhibitors. We review literature that
evaluates the in vivo potency of select CPDPs and attempt to
outline the areas of forthcoming progress, towards the development of CPDP-based
TLR inhibitors of pharmaceutical grade.
Collapse
Affiliation(s)
- Vladimir Y Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Artur Javmen
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Kumari T, Verma DP, Afshan T, Verma NK, Pant G, Ali M, Shukla PK, Mitra K, Ghosh JK. A Noncytotoxic Temporin L Analogue with In Vivo Antibacterial and Antiendotoxin Activities and a Nonmembrane-Lytic Mode of Action. ACS Infect Dis 2020; 6:2369-2385. [PMID: 32786286 DOI: 10.1021/acsinfecdis.0c00022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cytotoxic frog antimicrobial peptide Temporin L (TempL) is an attractive molecule for the design of lead antimicrobial agents due to its short size and versatile biological activities. However, noncytotoxic TempL variants with desirable biological activities have rarely been reported. TempL analogue Q3K,TempL is water-soluble and possesses a significant antiendotoxin property along with comparable cytotoxicity to TempL. A phenylalanine residue, located at the hydrophobic face of Q3K,TempL and the "d" position of its phenylalanine zipper sequence, was replaced with a cationic lysine residue. This analogue, Q3K,F8K,TempL, showed reduced hydrophobic moment and was noncytotoxic with lower antimicrobial activity. Interestingly, swapping between tryptophan at the fourth and serine at the sixth positions turned Q3K,F8K,TempL totally amphipathic as reflected by its helical wheel projection with clusters of hydrophobic and hydrophilic residues and the highest hydrophobic moment among these peptides. Surprisingly, this analogue, SW,Q3K,F8K,TempL, was as noncytotoxic as Q3K,F8K,TempL but showed augmented antimicrobial and antiendotoxin properties, comparable to that of TempL and Q3K,TempL. SW,Q3K,F8K,TempL exhibited appreciable survival of mice against P. aeruginosa infection and a lipopolysaccharide (LPS) challenge. Unlike TempL and Q3K,TempL, SW,Q3K,F8K,TempL adopted an unordered secondary structure in bacterial membrane mimetic lipid vesicles and did not permeabilize them or depolarize the bacterial membrane. Overall, the results demonstrate the design of a nontoxic TempL analogue that possesses clusters of hydrophobic and hydrophilic residues with impaired secondary structure and shows a nonmembrane-lytic mechanism and in vivo antiendotoxin and antimicrobial activities. This paradigm of design of antimicrobial peptide with clusters of hydrophobic and hydrophilic residues and high hydrophobic moment but low secondary structure could be attempted further.
Collapse
Affiliation(s)
- Tripti Kumari
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Devesh Pratap Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Tayyaba Afshan
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Neeraj Kumar Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Garima Pant
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Mehmood Ali
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - P. K. Shukla
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Jimut Kanti Ghosh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| |
Collapse
|
18
|
Suppressed nuclear factor-kappa B alleviates lipopolysaccharide-induced acute lung injury through downregulation of CXCR4 mediated by microRNA-194. Respir Res 2020; 21:144. [PMID: 32522221 PMCID: PMC7288420 DOI: 10.1186/s12931-020-01391-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/10/2020] [Indexed: 12/21/2022] Open
Abstract
Acute lung injury (ALI) is a highly lethal pulmonary disease that causes edema, hypoxemia and respiratory failure. Recent evidence indicates that nuclear factor-kappa B (NF-κB) plays a crucial role in ALI development. However, the regulatory mechanism of NF-κB on ALI remains enigmatic. In this study, we investigated potential molecular mechanism of NF-κB on ALI induced by lipopolysaccharide (LPS). BALB/c mice were subjected to intratracheal spraying of LPS to generate an ALI mode, with the activity of NF-κB in mice tissues being detected by enzyme linked immunosorbent assay (ELISA), and the number of inflammatory cells in bronchoalveolar lavage fluid being counted. Then, the macrophage cell line RAW264.7 exposed to LPS were treated with ammonium pyrrolidinedithiocarbamate (PDTC) (inhibitor of NF-κB), miR-194 mimic, or oe-chemokine receptor type 4 (CXCR4) separately or in combination. After that, ELISA and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used to detect the expression level of IL-1β, IL-6, TNF-α, miR-194 and CXCR4, respectively. In addition, the targeting relationship between miR-194 and CXCR4 was verified by dual-luciferase reporter gene assay. The dry/wet ratio of lung and the MPO activity were also measured to assess the inflammatory response in mice. Activation of NF-κB down-regulated the miR-194 expression in LPS-induced ALI. Overexpression of miR-194 alleviated LPS-induced ALI and reduced the expression of inflammatory factors IL-1β, IL-6 and TNF-α via targeting CXCR4. In LPS-induced ALI, NF-κB mediates the CXCR4 expression by inhibiting the expression of miR-194, thus promoting the inflammatory injury of lung.
Collapse
|
19
|
Gomes Dos Reis L, Traini D. Advances in the use of cell penetrating peptides for respiratory drug delivery. Expert Opin Drug Deliv 2020; 17:647-664. [PMID: 32138567 DOI: 10.1080/17425247.2020.1739646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Respiratory diseases are leading causes of death in the world, still inhalation therapies are the largest fail in drug development. There is an evident need to develop new therapies. Biomolecules represent apotential therapeutic agent in this regard, however their translation to the clinic is hindered by the lack of tools to efficiently deliver molecules. Cell penetrating peptides (CPPs) have arisen as apotential strategy for intracellular delivery that could theoretically enable the translation of new therapies.Areas covered: In this review, the use of CPPs as astrategy to deliver different molecules (cargoes) to treat lung-relateddiseases will be the focus. Abrief description of these molecules and the innovative methods in designing new CPPs is presented. The delivery of different cargoes (proteins, peptides, poorly soluble drugs and nucleic acids) using CPPs is discussed, focusing on benefits to treat different respiratory diseases like inflammatory disorders, cystic fibrosis and lung cancer.Expert opinion: The advantages of using CPPs to deliver biomolecules and poorly soluble drugs to the lungs is evident. This field has advanced in the past few years toward targeted intracellular delivery, although further studies are needed to fully understand its potential and limitations in vitro and in vivo.
Collapse
Affiliation(s)
- Larissa Gomes Dos Reis
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
20
|
Kang N, Shen W, Zhang Y, Su Z, Yang S, Liu Y, Xu Q. Anti-inflammatory and immune-modulatory properties of anemoside B4 isolated from Pulsatilla chinensis in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:152934. [PMID: 31454651 DOI: 10.1016/j.phymed.2019.152934] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/15/2019] [Accepted: 04/19/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Pulsatilla chinensis is commonly used for the treatment of cancers and inflammatory disorders in China. Our recent studies showed that anemoside B4, its major ingredient, possessed notable antioxidant and protected cisplatin-induced acute kidney injury in vivo. Furthermore, we found the protective effect might be involved its anti-inflammation activities. However, its anti-inflammatory mechanisms are not clear. PURPOSE In the present study, we extensively investigated the anti-inflammatory and immune-modulatory properties of anemoside B4 in vivo. METHODS To carry out this work, the xylene-induced ear edema and LPS-induced systemic inflammation of mice model was also used to evaluate the anti-inflammatory activity. Then, anti-inflammatory mechanism of anemoside B4 was further determined by pro-inflammatory cytokines production using enzyme-linked immunosorbent assay (ELISA) and nuclear factor-κ-gene binding (NF-κB) pathway activation by Western blot. At last, immuno-modulatory effects were observed by splenocyte proliferation assay, delayed type hypersensitivity assay (DTH) and T cell subtype assay in mice. RESULTS 12.5-50 mg/kg anemoside B4 significantly suppressed xylene-induced mice ear edema. Furthermore, it ameliorated LPS-induced kidney and lung inflammation damage, which inhibited pro-inflammatory response by NF-κB pathway in mice. In addition, anemoside B4 decreased CD4+/CD8+ ratio, inhibited splenic lymphocyte proliferation and decreased DNFB-induced changes of ear thickness. CONCLUSION From these data, it can be concluded that anemoside B4 presented anti-inflammatory and immune-modulatory activities in vivo, and potentially be a novel natural anti-inflammatory drug candidate for treating inflammatory disorder.
Collapse
Affiliation(s)
- Naixin Kang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Wenhua Shen
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; College of Pharmaceutical Science, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Yong Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Zhetong Su
- Sichuan Innovate Medical Technology Co. Ltd., Chengdu 610093, China
| | - Shilin Yang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China
| | - Yanli Liu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| | - Qiongming Xu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530000, China.
| |
Collapse
|
21
|
Transient receptor potential vanilloid 4 is a critical mediator in LPS mediated inflammation by mediating calcineurin/NFATc3 signaling. Biochem Biophys Res Commun 2019; 513:1005-1012. [PMID: 31005256 DOI: 10.1016/j.bbrc.2019.04.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/03/2019] [Indexed: 12/28/2022]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) ion channel is thought to be an essential component of inflammatory response. However, its role and mechanism in regulating acute lung injury (ALI) and macrophages activation are not well characterized. In our study, we observe that blockade of TRPV4 using GSK2193874 or HC-067047 greatly improve the pneumonedema, the lung pathologic changes, the up-regulation of proinflammatory cytokines and the neutrophil infiltration in LPS-induced lung injury. In vitro, knockdown of TRPV4 in macrophages reduces the levels of pro-inflammatory cytokines, ROS production, Ca2+ concentration in cytoplasma and the activation of calcineurin/NFATc3 signaling. Importantly, change of extracellular Ca2+ in culture medium prevents LPS-induced NFATc3 nuclear translocation, up-regulation of proinflammatory cytokines and ROS production in macrophages. Inhibition of calcineurin with cyclosporine A, FK506 down-regulates the levels of NFATc3 nuclear translocation and proinflammatory cytokines expression. Our results demonstrate that TRPV4-dependent Ca2+ influx contributes to LPS-induced macrophage activation by calcineurin-NFATc3 pathway.
Collapse
|
22
|
Levocetirizine Pretreatment Mitigates Lipopolysaccharide-Induced Lung Inflammation in Rats. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7019759. [PMID: 30186866 PMCID: PMC6110004 DOI: 10.1155/2018/7019759] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 01/02/2023]
Abstract
This research was conducted to investigate possible protective influences of levocetirizine, a nonsedating H1 antihistamine, against lipopolysaccharide (LPS)-induced lung injury in rats. Male Sprague Dawley rats received either levocetirizine (1 mg/kg/day, orally) or the vehicle of the drug (2 ml/kg/day, orally) for 1 week before a single IP injection of LPS (7.5 mg/kg). A group of normal rats served as control. The experiments were terminated 18 h after the LPS challenge. Serum C-reactive protein levels were determined. Moreover, total cell count, lactate dehydrogenase (LDH) activity, protein levels, and total NOx were evaluated in bronchoalveolar lavage fluid (BALF). Pulmonary edema was evaluated as the wet/dry lung weight ratio. Lung tissue homogenate was assessed for antioxidant/pro-oxidant status. BALF and lung tissue levels of tumor necrosis factor-α (TNF-α) were assessed. Lungs were examined for histological alterations. LPS-mediated lung injury was manifested by pulmonary edema, leukocyte infiltration, oxidative stress, and inflammation. Levocetirizine attenuated lung edema and mitigated the increases in BALF protein levels, LDH activity, and lung leukocyte recruitment in LPS-challenged rats. Additionally, TNF-α protein levels in BALF and lung tissue were diminished by levocetirizine administration. Levocetirizine also exhibited a potent antioxidant activity as indicated by a decrease in lung tissue levels of malondialdehyde and an enhancement of superoxide dismutase activity. Histological examination of lung tissues confirmed the beneficial effect of levocetirizine against LPS-induced histopathological alterations. In conclusion, levocetirizine may offer protection against lung tissue damage and inflammation in LPS-challenged rats.
Collapse
|
23
|
Yang J, Li S, Wang L, Du F, Zhou X, Song Q, Zhao J, Fang R. Ginsenoside Rg3 Attenuates Lipopolysaccharide-Induced Acute Lung Injury via MerTK-Dependent Activation of the PI3K/AKT/mTOR Pathway. Front Pharmacol 2018; 9:850. [PMID: 30116194 PMCID: PMC6082957 DOI: 10.3389/fphar.2018.00850] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical disease with high morbidity in both humans and animals. Ginsenoside Rg3, a type of traditional Chinese medicine extracted from ginseng, is widely used to cure many inflammation-related diseases. However, the specific molecular mechanism of the effects of ginsenoside Rg3 on inflammation has rarely been reported. Thus, we established a mouse model of lipopolysaccharide (LPS)-induced ALI to investigate the immune protective effects of ginsenoside Rg3 and explore its molecular mechanism. In wild type (WT) mice, we found that ginsenoside Rg3 treatment significantly mitigated pathological damages and reduced myeloperoxidase (MPO) activity as well as the production of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6); furthermore, the production of anti-inflammatory mediators interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), polarization of M2 macrophages and expression levels of the phosphorylation of phosphatidylinositol 3-hydroxy kinase (PI3K), protein kinase B (PKB, also known as AKT), mammalian target of rapamycin (mTOR) and Mer receptor tyrosine kinase (MerTK) were promoted. However, there were no significant differences with regards to the pathological damage, MPO levels, inflammatory cytokine levels, and protein expression levels of the phosphorylation of PI3K, AKT and mTOR between the LPS treatment group and ginsenoside Rg3 group in MerTK-/- mice. Taken together, the present study demonstrated that ginsenoside Rg3 could attenuate LPS-induced ALI by decreasing the levels of pro-inflammatory mediators and increasing the production of anti-inflammatory cytokines. These processes were mediated through MerTK-dependent activation of its downstream the PI3K/AKT/mTOR pathway. These findings identified a new site of the specific anti-inflammatory mechanism of ginsenoside Rg3.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Luyao Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fen Du
- Hubei Center for Animal Diseases Control and Prevention, Wuhan, China
| | - Xiaoliu Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiqi Song
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
24
|
Wang C, Meng Y, Wang Y, Jiang Z, Xu M, Bo L, Deng X. Ouabain Protects Mice Against Lipopolysaccharide-Induced Acute Lung Injury. Med Sci Monit 2018; 24:4455-4464. [PMID: 29953424 PMCID: PMC6053945 DOI: 10.12659/msm.908627] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Ouabain, an inhibitor of Na+/K+-ATPase, is a type of endogenous hormone synthesized in the adrenal cortex and hypothalamus. Previous studies found that ouabain potently inhibited inflammatory reactions and regulated immunological processes. Our present study aimed to investigate the therapeutic role of ouabain on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Material/Methods Ouabain (0.1 mg/kg) or vehicles were intraperitoneally injected into male C57BL/6J mice once a day for 3 consecutive days. One hour after the last injection of ouabain, LPS (5 mg/kg) was administrated through intranasal instillation to induce ALI. 6 hours and 24 hours later, bronchoalveolar lavage fluid (BALF) and lung tissues were harvested to detect the protective effects of ouabain, including protein concentration, inflammation cell counts, lung wet-to-dry ratio, and lung damage. Results The results showed that ouabain attenuated LPS-induced ALI in mice, which was indicated by alleviated pathological changes, downregulated TNF-α, IL-1β, and IL-6 production, inhibited neutrophils infiltration and macrophages, and ameliorated pulmonary edema and permeability. Further results found the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways were suppressed by ouabain in LPS-induced ALI. Conclusions These results suggest that ouabain negatively modulates the severity of LPS-induced ALI.
Collapse
Affiliation(s)
- Changli Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Yan Meng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Yuanyuan Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland).,Department of Anesthesiology, Women and Children's Health Care Hospital of Linyi City, Linyi, Shandong, China (mainland)
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Mengda Xu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| |
Collapse
|
25
|
Javmen A, Szmacinski H, Lakowicz JR, Toshchakov VY. Blocking TIR Domain Interactions in TLR9 Signaling. THE JOURNAL OF IMMUNOLOGY 2018; 201:995-1006. [PMID: 29914886 DOI: 10.4049/jimmunol.1800194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/24/2018] [Indexed: 01/07/2023]
Abstract
Interaction of TLR9 with ligands activates NF-κB, leading to proinflammatory cytokine production. Excessive TLR activation is a pathogenic factor for inflammatory diseases. This study has examined cell-permeating decoy peptides (CPDPs) derived from the TLR9 Toll/IL-1R resistance (TIR) domain. CPDP 9R34, which included AB loop, β-strand B, and N-terminal BB loop residues, inhibited TLR9 signaling most potently. CPDPs derived from α-helices C, D, and E (i.e., 9R6, 9R9, and 9R11) also inhibited TLR9-induced cytokines but were less potent than 9R34. 9R34 did not inhibit TLR2/1, TLR4, or TLR7 signaling. The N-terminal deletion modification of 9R34, 9R34-ΔN, inhibited TLR9 as potently as the full length 9R34. Binding of 9R34-ΔN to TIR domains was studied using cell-based Förster resonance energy transfer/fluorescence lifetime imaging approach. Cy3-labeled 9R34-ΔN dose-dependently decreased fluorescence lifetime of TLR9 TIR-Cerulean (Cer) fusion protein. Cy3-9R34-ΔN also bound TIRAP TIR, albeit with a lesser affinity, but not MyD88 TIR, whereas CPDP from the opposite TIR surface, 9R11, bound both adapters and TLR9. i.p. administration of 9R34-ΔN suppressed oligonucleotide-induced systemic cytokines and lethality in mice. This study identifies a potent, TLR9-specific CPDP that targets both receptor dimerization and adapter recruitment. Location of TIR segments that represent inhibitory CPDPs suggests that TIR domains of TLRs and TLR adapters interact through structurally homologous surfaces within primary receptor complex, leading to formation of a double-stranded, filamentous structure. In the presence of TIRAP and MyD88, primary complex can elongate bidirectionally, from two opposite ends, whereas in TIRAP-deficient cells, elongation is unidirectional, only through the αE side.
Collapse
Affiliation(s)
- Artur Javmen
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Henryk Szmacinski
- Center for Fluorescence Spectroscopy, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Joseph R Lakowicz
- Center for Fluorescence Spectroscopy, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Vladimir Y Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| |
Collapse
|
26
|
Zhao G, Yang C, Yang J, Liu P, Jiang K, Shaukat A, Wu H, Deng G. Placental exosome-mediated Bta-miR-499-Lin28B/let-7 axis regulates inflammatory bias during early pregnancy. Cell Death Dis 2018; 9:704. [PMID: 29899331 PMCID: PMC5999645 DOI: 10.1038/s41419-018-0713-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022]
Abstract
Abnormal inflammatory bias in the maternal-fetal interface leads to reproductive failure in mammals. Placental exosomes are involved in maternal-fetal communication during pregnancy. However, whether the placenta or fetus is involved in regulating the balance of uterine local inflammation through exosomes remains unclear, and the mechanism must be further explored. Here we demonstrated that placenta-specific exosomes are abundant in the peripheral blood of dairy cows during early pregnancy and selectively load miRNAs, such as bta-miR-499. In vitro, placental exosome-derived bta-miR-499 inhibits the activation of NF-κB via the Lin28B/let-7 axis, thus repressing LPS-induced inflammation in bovine endometrial epithelial (BEND) cells. Subsequently, inhibition of mmu-miR-499 leads to an impaired balance of inflammation at the maternal-fetal interface in vivo, resulting in an increased risk of pregnancy failure due to placental loss and fetal growth restriction. Thus, our data demonstrate that placental exosomal miR-499 may be a critical immune regulator in the regulation of the inflammation balance at the maternal-fetal interface in the early gestation of dairy cows and other mammals.
Collapse
Affiliation(s)
- Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Chao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Pei Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
27
|
Tang F, Fan K, Wang K, Bian C. Atractylodin attenuates lipopolysaccharide-induced acute lung injury by inhibiting NLRP3 inflammasome and TLR4 pathways. J Pharmacol Sci 2018; 136:203-211. [DOI: 10.1016/j.jphs.2017.11.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/16/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022] Open
|
28
|
Zhang F, Fan D, Mo XN. Prohibitin and the extracellular matrix are upregulated in murine alveolar epithelial cells with LPS‑induced acute injury. Mol Med Rep 2018; 17:7769-7773. [PMID: 29620269 DOI: 10.3892/mmr.2018.8808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 11/23/2017] [Indexed: 11/05/2022] Open
Abstract
Inflammation of epithelial and endothelial cells accelerates the progress of acute lung injury (ALI), and pulmonary fibrosis is the leading cause of mortality in patients with acute respiratory distress syndrome. Interleukin‑6 (IL‑6) is a pleiotropic cytokine implicated in the pathogenesis of a number of immune‑mediated disorders, and is involved in pulmonary fibrosis. Prohibitin (PHB) is a highly conserved protein implicated in various cellular functions, including proliferation, apoptosis, tumor suppression, transcription and mitochondrial protein folding. PHB was identified to be associated with a variety of pulmonary diseases, including pulmonary fibrosis. Based on the lipopolysaccharide (LPS)‑induced cell model of ALI, the present study examined the expression of PHB and the extracellular matrix (ECM) in the process of pulmonary inflammation. MLE‑12 cells were divided into 2 groups: The control group was administered sterile PBS; the treatment group was administered 500 ng/ml LPS for 12 h. The mRNA expression of IL‑6 in the treatment group was significantly upregulated compared with the control group (P<0.05). The protein expression of IL‑6 in the treatment group was markedly increased compared with the control group (P<0.05). ECM components, including collagen‑IV and fibronectin, in the treatment group were markedly increased when compared with the control group (P<0.05). The mRNA and protein expression levels of PHB1 and PHB2 were significantly upregulated following treatment with LPS (both P<0.05). The present study identified that PHB and ECM component levels increased in the LPS‑induced ALI cell model, and further investigations may be performed to verify the detailed mechanism.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Respiratory Medicine, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Dejun Fan
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Xiao-Neng Mo
- Department of Respiratory Medicine, The Sixth Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
29
|
Lin SQ, Zhou ZL, Li CY. Cyprotuoside C and Cyprotuoside D, Two New Cycloartane Glycosides from the Rhizomes of Cyperus rotundus. Chem Pharm Bull (Tokyo) 2018; 66:96-100. [PMID: 29118309 DOI: 10.1248/cpb.c17-00608] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyprotuoside C (1) and cyprotuoside D (2), two new cycloartane glycosides were isolated from the ethanol extract of the rhizomes of Cyperus rotundus. Their structures were identified as 24R-9,10-seco-cycloartan-1(10),9(11)-dien-3β,7β,24,25-tetraol 3-O-β-D-xylopyranosyl-(1→4)-[α-L-arabinopyranosyl-(1→6)]-β-D-glucopyranosyl-25-O-β-D-glucuronide (1) and 9,10-seco-cycloartan-1(10),9(11),23(24)-trien-3β,7β,25-triol 3-O-β-D-xylopyranosyl-(1→4)-{α-L-arabinopyranosyl-(1→6)-[α-L-rhamnopyranosyl-(1→2)]}-β-D-glucopyranosyl-25-O-β-D-glucuronide (2) by spectroscopic methods.
Collapse
Affiliation(s)
- San-Qing Lin
- School of Chemistry and Chemical Engineering, Lingnan Normal University
| | - Zhong-Liu Zhou
- School of Chemistry and Chemical Engineering, Lingnan Normal University
| | - Chun-Yan Li
- School of Chemistry and Chemical Engineering, Lingnan Normal University
| |
Collapse
|