Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.518
Filter
1.
J Ethnopharmacol ; 336: 118661, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39159837

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Shuangdan Jiedu Decoction (SJD) is a formula composed of six Chinese herbs with heat-removing and detoxifying, antibacterial, and anti-inflammatory effects, which is clinically used in the therapy of various inflammatory diseases of the lungs including COVID-19, but the therapeutic material basis of its action as well as its molecular mechanism are still unclear. AIM OF THE STUDY: The study attempted to determine the therapeutic effect of SJD on LPS-induced acute lung injury (ALI), as well as to investigate its mechanism of action and assess its therapeutic potential for the cure of inflammation-related diseases in the clinical setting. MATERIALS AND METHODS: We established an ALI model by tracheal drip LPS, and after the administration of SJD, we collected the bronchoalveolar lavage fluid (BALF) and lung tissues of mice and examined the expression of inflammatory factors in them. In addition, we evaluated the effects of SJD on the cyclic guanosine monophosphate-adenosine monophosphate synthase -stimulator of interferon genes (cGAS-STING) and inflammasome by immunoblotting and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS: We demonstrated that SJD was effective in alleviating LPS-induced ALI by suppressing the levels of pro-inflammatory cytokines in the BALF, improving the level of lung histopathology and the number of neutrophils, as well as decreasing the inflammatory factor-associated gene expression. Importantly, we found that SJD could inhibit multiple stimulus-driven activation of cGAS-STING and inflammasome. Further studies showed that the Chinese herbal medicines in SJD had no influence on the cGAS-STING pathway and inflammasome alone at the formulated dose. By increasing the concentration of these herbs, we observed inhibitory effects on the cGAS-STING pathway and inflammasome, and the effect exerted was maximal when the six herbs were combined, indicating that the synergistic effects among these herbs plays a crucial role in the anti-inflammatory effects of SJD. CONCLUSIONS: Our research demonstrated that SJD has a favorable protective effect against ALI, and its mechanism of effect may be associated with the synergistic effect exerted between six Chinese medicines to inhibit the cGAS-STING and inflammasome abnormal activation. These results are favorable for the wide application of SJD in the clinic as well as for the development of drugs for ALI from herbal formulas.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Inflammasomes , Lipopolysaccharides , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Lipopolysaccharides/toxicity , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Nucleotidyltransferases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Male , Signal Transduction/drug effects , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Lung/drug effects , Lung/pathology , Lung/metabolism , Bronchoalveolar Lavage Fluid/cytology
2.
J Ethnopharmacol ; 336: 118730, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181280

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Acute lung injury (ALI) can lead to respiratory failure and even death. KAT2A is a key target to suppress the development of inflammation. A herb, perilla frutescens, is an effective treatment for pulmonary inflammatory diseases with anti-inflammatory effects; however, its mechanism of action remains unclear. AIM OF THE STUDY: The purpose of this study was to investigate the therapeutic effect and underlying mechanism of perilla frutescens leaf extracts (PLE), in the treatment of ALI by focusing on its ability to treat inflammation. MATERIALS AND METHODS: In vivo and in vitro models of ALI induced by LPS. Respiratory function, histopathological changes of lung, and BEAS-2B cells damage were assessed upon PLE. This effect is also tested under conditions of KAT2A over expression and KAT2A silencing. RESULTS: PLE significantly attenuated LPS-induced histopathological changes in the lungs, improved respiratory function, and increased survival rate from LPS stimuation background in mice. PLE remarkably suppressed the phosphorylation of STAT3, AKT, ERK (1/2) and the release of cytokines (IL-6, TNF-α, and IL-1ß) induced by LPS via inhibiting the expression of KAT2A. CONCLUSIONS: PLE has a dose-dependent anti-inflammatory effect by inhibiting KAT2A expression to suppress LPS-induced ALI n mice. Our study expands the clinical indications of the traditional medicine PLE and provide a theoretical basis for clinical use of acute lung injury.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Perilla frutescens , Plant Extracts , Plant Leaves , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Perilla frutescens/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Male , Mice , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Cell Line , Mice, Inbred C57BL , Lung/drug effects , Lung/pathology , Lung/metabolism , Disease Models, Animal
3.
J Ethnopharmacol ; 336: 118699, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181290

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Acute lung injury (ALI) is a serious health-threatening syndrome of intense inflammatory response in the lungs, with progression leading to acute respiratory distress syndrome (ARDS). Dachengqi decoction dispensing granule (DDG) has a pulmonary protective role, but its potential modulatory mechanism to alleviate ALI needs further excavation. AIM OF THE STUDY: This study aims to investigate the effect and potential mechanism of DDG on lipopolysaccharide (LPS)-induced ALI models in vivo and in vitro. MATERIALS AND METHODS: LPS-treated Balb/c mice and BEAS-2B cells were used to construct in vivo and in vitro ALI models, respectively. Hematoxylin-eosin (HE), Wet weight/Dry weight (W/D) calculation of lung tissue, and total protein and Lactic dehydrogenase (LDH) assays in BALF were performed to assess the extent of lung tissue injury and pulmonary edema. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), and interleukin-18 (IL-18) in BALF, serum, and cell supernatant. The qRT-PCR was used to detect inflammatory factors, Z-DNA binding protein 1 (ZBP1), and receptor-interacting protein kinase 1 (RIPK1) expression in lung tissues and BEAS-2B cells. Double immunofluorescence staining and co-immunoprecipitation were used to detect the relative expression and co-localization of ZBP1 and RIPK1. The effects of LPS and DDG on BEAS-2B cell activity were detected by Cell Counting Kit-8 (CCK-8). Western blot (WB) was performed to analyze the expression of PANoptosis-related proteins in lung tissues and BEAS-2B cells. RESULTS: In vivo, DDG pretreatment could dose-dependently improve the pathological changes of lung tissue in ALI mice, and reduce the W/D ratio of lung, total protein concentration, and LDH content in BALF. In vitro, DDG reversed the inhibitory effect of LPS on BEAS-2B cell viability. Meanwhile, DDG significantly reduced the levels of inflammatory factors in vitro and in vivo. In addition, DDG could inhibit the expression levels of PANoptosis-related proteins, especially the upstream key regulatory molecules ZBP1 and RIPK1. CONCLUSION: DDG could inhibit excessive inflammation and PANoptosis to alleviate LPS-induced ALI, thus possessing good anti-inflammatory and lung-protective effects. This study establishes a theoretical basis for the further development of DDG and provides a new prospect for ALI treatment by targeting PANoptosis.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Mice, Inbred BALB C , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Lipopolysaccharides/toxicity , Humans , Male , Mice , Cell Line , Lung/drug effects , Lung/pathology , Lung/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Plant Extracts/pharmacology , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use
4.
Can Respir J ; 2024: 1068326, 2024.
Article in English | MEDLINE | ID: mdl-39268525

ABSTRACT

Sepsis is a systemic inflammatory reaction syndrome caused by infections. Acute lung injury (ALI) occurs first and most frequently in patients with sepsis. Gentiopicroside (GPS), which originates mostly from Gentiana, is classified as a secoiridoid glycosides. Terpenoid glycosides have various biological effects, including liver protection, blood glucose and cholesterol level management, and anti-inflammatory and antitumor effects. However, presently, the biochemical foundation and mechanism of the anti-inflammatory effects of GPS in sepsis-induced ALI have not been explained. In the present study, we established a rat model of sepsis ALI induced by cecal ligation and puncture. This enables us to observe the effects of GPS therapy, which significantly reduced the inflammatory response (TNF-α, IL-1ß, and IL-6), nitrogen stress, oxidative stress, and severity of ALI at both the whole animal and molecular levels. In addition, GPS ameliorates LPS-induced ALI via regulation of inflammatory response and cell proptosis in BEAS-2B. This study provides a theoretical basis for treating sepsis-induced ALI with GPS.


Subject(s)
Acute Lung Injury , Iridoid Glucosides , Sepsis , Animals , Sepsis/complications , Sepsis/drug therapy , Iridoid Glucosides/pharmacology , Iridoid Glucosides/therapeutic use , Acute Lung Injury/etiology , Acute Lung Injury/drug therapy , Rats , Male , Rats, Sprague-Dawley , Disease Models, Animal , Oxidative Stress/drug effects , Inflammation/drug therapy
5.
Zhongguo Zhong Yao Za Zhi ; 49(16): 4329-4337, 2024 Aug.
Article in Chinese | MEDLINE | ID: mdl-39307770

ABSTRACT

This study aims to investigate the mechanism of Xuanbai Chengqi Decoction in treating acute lung injury(ALI) based on network pharmacology and animal experiments. The potential targets and signaling pathways of Xuanbai Chengqi Decoction in regulating ALI were predicted by network pharmacology. The rat model of ALI was constructed and administrated with different doses of Xuanbai Chengqi Decoction. The pathological changes in the lung tissue of rats were observed by hematoxylin-eosin(HE) staining. The levels of interleukin-6(IL-6), interleukin-1ß(IL-1ß), and tumor necrosis factor-α(TNF-α) in the peripheral blood were measured by enzyme-linked immunosorbent assay(ELISA). The mRNA and protein levels of factors in the phosphatidylinositol 3-kinase(PI3K)/protein kinase B(Akt)/mammalian target of rapamycin(mTOR) signaling pathway were determined by quantitative real-time PCR(qPCR) and Western blot, respectively. A total of 52 compounds from Xuanbai Chengqi Decoction were predicted to be involved in the treatment of ALI, including ß-sitosterol, emodin, stigmasterol, glabridin, and aloe-emodin, which corresponded to 112 targets,and 4 723 targets of ALI were predicted. The compounds and ALI shared 94 common targets. The key targets included TNF, IL-1ß,prostaglandin-endoperoxide synthase 2(PTGS2), and tumor protein 53(TP53). Lipids and atherosclerosis, p53 signaling pathway,IL-17 signaling pathway, and PI3K/Akt signaling pathway were mainly involved in the treatment. Animal experiments showed that compared with the model group, Xuanbai Chengqi Decoction alleviated the pathological changes in the lung tissue, lowered the serum levels of IL-6, IL-1ß, and TNF-α, down-regulated the mRNA and protein levels of PI3K, Akt, and mTOR, and reduced the p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR ratios in ALI rats. The results showed that Xuanbai Chengqi Decoction exerted its therapeutic effects on ALI via multiple components, targets, and pathways. Meanwhile, Xuanbai Chengqi Decoction may reduce the inflammation and attenuate the lung injuries of ALI rats by inhibiting the PI3K/Akt/mTOR signaling pathway.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Interleukin-1beta , Network Pharmacology , Rats, Sprague-Dawley , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Rats , Signal Transduction/drug effects , Male , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Humans , Lung/drug effects , Lung/metabolism
6.
Malar J ; 23(1): 285, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300444

ABSTRACT

BACKGROUND: Severe malaria can cause respiratory symptoms, which may lead to malaria-acute lung injury (MA-ALI) due to inflammation and damage to the blood-gas barrier. Patients with severe malaria also often present thrombocytopenia, and the use of acetylsalicylic acid (ASA), a commonly used non-steroidal anti-inflammatory drug with immunomodulatory and antiplatelet effects, may pose a risk in regions where malaria is endemic. Thus, this study aimed to investigate the systemic impact of ASA and dihydroartemisinin (DHA) on ALI induced in mice by Plasmodium berghei NK65 (PbNK65). METHODS: C57BL/6 mice were randomly divided into control (C) and PbNK65 infected groups and were inoculated with uninfected or 104 infected erythrocytes, respectively. Then, the animals were treated with DHA (3 mg/kg) or vehicle (DMSO) at the 8-day post-infection (dpi) for 7 days and with ASA (100 mg/kg, single dose), and analyses were performed at 9 or 15 dpi. Lung mechanics were performed, and lungs were collected for oedema evaluation and histological analyses. RESULTS: PbNK65 infection led to lung oedema, as well as increased lung static elastance (Est, L), resistive (ΔP1, L) and viscoelastic (ΔP2, L) pressures, percentage of mononuclear cells, inflammatory infiltrate, hemorrhage, alveolar oedema, and alveolar thickening septum at 9 dpi. Mice that received DHA or DHA + ASA had an increase in Est, L, and CD36 expression on inflammatory monocytes and higher protein content on bronchoalveolar fluid (BALF). However, only the DHA-treated group presented a percentage of inflammatory monocytes similar to the control group and a decrease in ΔP1, L and ΔP2, L compared to Pb + DMSO. Also, combined treatment with DHA + ASA led to an impairment in diffuse alveolar damage score and lung function at 9 dpi. CONCLUSIONS: Therapy with ASA maintained lung morpho-functional impairment triggered by PbNK65 infection, leading to a large influx of inflammatory monocytes to the lung tissue. Based on its deleterious effects in experimental MA-ALI, ASA administration or its treatment maintenance might be carefully reconsidered and further investigated in human malaria cases.


Subject(s)
Acute Lung Injury , Antimalarials , Artemisinins , Aspirin , Lung , Malaria , Mice, Inbred C57BL , Plasmodium berghei , Animals , Artemisinins/pharmacology , Acute Lung Injury/drug therapy , Acute Lung Injury/parasitology , Aspirin/pharmacology , Aspirin/administration & dosage , Malaria/drug therapy , Malaria/complications , Mice , Antimalarials/pharmacology , Plasmodium berghei/drug effects , Lung/pathology , Lung/drug effects , Drug Therapy, Combination , Disease Models, Animal , Male , Respiratory Function Tests
7.
PLoS One ; 19(9): e0309721, 2024.
Article in English | MEDLINE | ID: mdl-39302999

ABSTRACT

The purpose of this paper was to study in vitro atomization properties of the self-developed sodium sivelestat for inhalation, evaluate the feasibility of this preparation as an aerosol inhalation, and provide the guidance for the following animal administration experiment. Firstly, in order to ensure accurate, uniform and stable doses of the self-developed product after administration, its atomization performance was analyzed through the testing of fine particle mass and the total emitted dose, and the results of its atomization parameters meet the requirement of inhalation. Next, Atomization characteristics of two commonly used nebulizers, air compressed nebulizer and mesh nebulizer, were studied and compared. The results showed that mesh atomizers have a smaller and more uniform particle size distribution. And then, the experiment of acute lung injury induced by aerosol inhalation of lipopolysaccharide in mice was used to test the therapeutic effect of our self-developed formulation, and compared with the positive control (sodium sivelestat for injection). The results showed that inhalation had a lower concentration and was equally effective than injection of sodium sivelestat. All the results support that the self-developed sodium sivelestat can be used as an aerosol inhaled drug.


Subject(s)
Aerosols , Glycine , Nebulizers and Vaporizers , Particle Size , Sulfonamides , Animals , Administration, Inhalation , Mice , Sulfonamides/administration & dosage , Glycine/analogs & derivatives , Glycine/administration & dosage , Glycine/chemistry , Acute Lung Injury/drug therapy , Male
8.
Respir Res ; 25(1): 354, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39342264

ABSTRACT

BACKGROUND: Exposure to a hypobaric hypoxic environment at high altitudes can lead to lung injury. In this study, we aimed to determine whether curcumin (Cur) could improve lung barrier function and protect against high-altitude-associated acute lung injury. METHODS: Two hundred healthy rats were randomly divided into standard control, high-altitude control (HC), salidroside (40 mg/kg, positive control), and Cur (200 mg/kg) groups. Each group was further divided into five subgroups. Basic vital signs, lung injury histopathology, routine blood parameters, plasma lactate level, and arterial blood gas indicators were evaluated. Protein and inflammatory factor (tumor necrosis factor α (TNF-α), interleukin [IL]-1ß, IL-6, and IL-10) concentrations in bronchoalveolar lavage fluid (BALF) were determined using the bicinchoninic acid method and enzyme-linked immunosorbent assay, respectively. Inflammation-related and lung barrier function-related proteins were analyzed using immunoblotting. RESULTS: Cur improved blood routine indicators such as hemoglobin and hematocrit and reduced the BALF protein content and TNF-α, IL-1ß, and IL-6 levels compared with those in the HC group. It increased IL-10 levels and reduced pulmonary capillary congestion, alveolar hemorrhage, and the degree of pulmonary interstitial edema. It increased oxygen partial pressure, oxygen saturation, carbonic acid hydrogen radical, and base excess levels, and the expression of zonula occludens 1, occludin, claudin-4, and reduced carbon dioxide partial pressure, plasma lactic acid, and the expression of phospho-nuclear factor kappa. CONCLUSIONS: Exposure to a high-altitude environment for 48 h resulted in severe lung injury in rats. Cur improved lung barrier function and alleviated acute lung injury in rats at high altitudes.


Subject(s)
Acute Lung Injury , Altitude Sickness , Curcumin , Rats, Sprague-Dawley , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Rats , Male , Curcumin/pharmacology , Curcumin/therapeutic use , Altitude Sickness/drug therapy , Altitude Sickness/metabolism , Altitude Sickness/complications , Altitude Sickness/physiopathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Altitude , Inflammation Mediators/metabolism , Bronchoalveolar Lavage Fluid , Cytokines/metabolism
9.
Clin Respir J ; 18(9): e70004, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39313818

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) is a critical and lethal medical condition. This syndrome is characterized by an imbalance in the body's oxidation stress and inflammation. Linoleic acid (LA), a polyunsaturated fatty acid, has been extensively studied for its potential health benefits, including anti-inflammatory and antioxidant activities. However, the therapeutic effects of LA on ALI remain unexplored. METHODS: Lipopolysaccharide (LPS), found in gram-negative bacteria's outer membrane, was intraperitoneally injected to induce ALI in mice. In vitro model was established by LPS stimulation of mouse lung epithelial 12 (MLE-12) cells. RESULTS: LA treatment demonstrated a significant amelioration in LPS-induced hypothermia, poor state, and pulmonary injury in mice. LA treatment resulted in a reduction in the concentration of bronchoalveolar lavage fluid (BALF) protein and an increase in myeloperoxidase (MPO) activity in LPS-induced mice. LA treatment reduced the generation of white blood cells. LA treatment reduced cell-free (cfDNA) release and promote adenosine triphosphate (ATP) production. LA increased the levels of superoxide dismutase (SOD) and glutathione (GSH) but decreased the production of malondialdehyde (MDA). LA treatment enhanced mitochondrial membrane potential. LA attenuated LPS-induced elevations of inflammatory cytokines in both mice and cells. Additionally, LA exerted its protective effect against LPS-induced damage through activation of the peroxisome proliferator-activated receptor γ coactivator l alpha (PGC-1α)/nuclear respiratory factor 1 (NRF1)/transcription factor A of the mitochondrion (TFAM) pathway. CONCLUSION: LA may reduce inflammation and stimulate mitochondrial biogenesis in ALI mice and MLE-12 cells.


Subject(s)
Acute Lung Injury , Disease Models, Animal , Linoleic Acid , Lipopolysaccharides , Organelle Biogenesis , Animals , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/drug therapy , Mice , Linoleic Acid/metabolism , Linoleic Acid/pharmacology , Male , Oxidative Stress/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Bronchoalveolar Lavage Fluid
10.
J Pharmacol Sci ; 156(3): 188-197, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39313277

ABSTRACT

Acute lung injury (ALI) is closely related to high mortality in severe acute pancreatitis (SAP). This study unveils the therapeutic effect and mechanism of miR-217-5p on SAP-associated ALI. The miR-217-5p RNA expression was significantly up-regulated in lipopolysaccharide (LPS)-stimulated primary rat alveolar epithelial type II cells (AEC II) and sodium taurocholate-treated pancreas and lung in SAP rats. miR-217 inhibition protected AEC II from LPS-induced damage by inhibiting apoptosis and reducing the TNF-α, IL-6, and ROS levels. miR-217 inhibition suppressed apoptosis and alleviated mitochondrial damage through mitochondria-mediated apoptotic pathway in vitro. Sirt1 is a direct target of miR-217-5p. Dual-luciferase reporter assay confirmed the binding of miR-217-5p to Sirt1 mRNA 3'-UTR. The rescue experiment identified that the anti-apoptotic, anti-inflammatory, and anti-oxidative effects of miR-217 inhibition were mediated by Sirt1 in vitro. Emodin (EMO) protected AEC II from LPS-induced damage and alleviated pancreatic and lung tissue injuries. EMO exerted similar effects as miR-217 inhibition in vitro and in vivo. The effects of EMO were abolished by miR-217 overexpression. In conclusion, miR-217-5p inhibition exerts protective effects on SAP-ALI in vitro and in vivo by repressing apoptosis, inflammation, and oxidative stress through Sirt1 activation. EMO protects against lung injuries in SAP-associated ALI rats through miR-217-5p/Sirt1 axis.


Subject(s)
Acute Lung Injury , Apoptosis , Emodin , MicroRNAs , Pancreatitis , Rats, Sprague-Dawley , Sirtuin 1 , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Emodin/pharmacology , Emodin/therapeutic use , Male , Pancreatitis/drug therapy , Pancreatitis/metabolism , Pancreatitis/genetics , Pancreatitis/chemically induced , Apoptosis/drug effects , Apoptosis/genetics , Lipopolysaccharides/adverse effects , Rats , Cells, Cultured , Acute Disease , Disease Models, Animal
11.
Phytomedicine ; 134: 155985, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39236558

ABSTRACT

BACKGROUND: Hua-Shi-Bai-Du decoction (HSBD) exerts significant effects on the prevention and treatment of COVID-19 in China. The activation of the NLRP3 inflammasome of macrophages plays a vital role in COVID-19 pathology. However, no previous studies have focused on this pathological process to explore the effect of HSBD. PURPOSE: Our aim is to uncover the effect of HSBD on NLRP3 inflammasome activation and the underlying mechanisms. METHODS: The NLRP3-activated J774A.1 cells primed by LPS and activated by nigericin/ATP/MSU were used to evaluate NLRP3 activation in vitro. ASC oligomerization and speck formation were assessed by western blot and immunofluorescence imaging. Intracellular K+ levels were determined by the colorimetric assay. Mitochondrial ROS (mtROS) level was detected by the flow cytometry and the fluorescence spectrophotometry. The intracellular cAMP level was determined by chemiluminescence method and ELISA, while phosphodiesterase (PDE) activity was measured using the fluorescent substrate MANT-cAMP. siRNA was applied to knockdown PDE4B. Two in vivo mouse models, MSU-induced peritonitis and LPS-induced acute lung injury (ALI), were used to evaluate the effects of HSBD on IL-1ß and other inflammatory cytokines. Pathological changes in lung tissue were observed by histopathological examination. RESULTS: HSBD not only decreased supernatant IL-1ß, caspase-1 p20, and cleaved gasdermin D (GSDMD) in NLRP3-activated J774A.1 cells, but also reduced IL-1ß in the peritoneal lavage fluid of mice with MSU-induced peritonitis, demonstrating the suppressive effect on NLRP3 inflammasome activation. The mechanism study showed that HSBD blocked ASC oligomerization and speck formation without affecting K+ efflux or mtROS production. Furthermore, it prevented the decrease of intracellular cAMP by inhibiting PDE4B activity. And in the PDE4B-deficient cells, its suppressive effect on IL-1ß release was abolished. In LPS-induced ALI mice, oral administration of HSBD decreased several proinflammatory cytokines (IL-1ß, IL-6, TNF-α, and CXCL-1) and attenuated the pathological damage to the lung. CONCLUSION: HSBD suppresses the activation of NLRP3 inflammasome by inhibiting PDE4B activity to counteract the decrease of intracellular cAMP, thereby blocking ASC oligomerization in macrophages. Our findings may provide new insight into the clinical effets of HSBD for the treatment of COVID-19.


Subject(s)
Acute Lung Injury , Cyclic Nucleotide Phosphodiesterases, Type 4 , Drugs, Chinese Herbal , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Lung Injury/drug therapy , Mice , Inflammasomes/metabolism , Inflammasomes/drug effects , Drugs, Chinese Herbal/pharmacology , Macrophages/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Male , Disease Models, Animal , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Cell Line , COVID-19 , Interleukin-1beta/metabolism , Lipopolysaccharides
12.
BMC Complement Med Ther ; 24(1): 334, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272057

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) as one kind of acute pulmonary inflammatory disorder, manifests primarily as damage to alveolar epithelial cells and microvascular endothelial cells. Activation of the complement system is a common pathological mechanism in ALI induced by diverse factors, with the complement alternative pathway assuming a pivotal role. Baicalin, a flavonoid derived from the root of Scutellaria baicalensis Georgi, exhibits noteworthy biological activities. The present study attempted the interventional effects and underlying mechanisms of baicalin in microangiopathy in ALI induced by complement alternative pathway activation. METHODS: Activation of the complement alternative pathway by cobra venom factor (CVF). HMEC cells were pretreated with baicalin and then exposed to complement activation products. The expression of inflammatory mediators was detected by ELISA, and the intranuclear transcriptional activity of NF-κB was assessed by a dual fluorescent kinase reporter gene assay kit. Before establishing the ALI mouse model, baicalin or PDTC was gavaged for 7 d. CVF was injected into the tail vein to establish the ALI model. The levels of inflammatory mediators in BALF and serum were determined by ELISA. HE staining and immunohistochemistry evaluated pathological changes, complement activation product deposition, and NF-κB p65 phosphorylation in lung tissue. RESULTS: Baicalin reduced complement alternative activation product-induced expression of HMEC cells adhesion molecules (ICAM-1, VCAM-1, E-selectin) and cytokines (IL-6, TNF-α) as well as upregulation of NF-κB intranuclear transcriptional activity. Baicalin intervention reduced the number of inflammatory cells and protein content in the BALF and decreased the levels of IL-6, TNF-α, and ICAM-1 in serum and IL-6, TNF-α, ICAM-1, and P-selectin in BLAF. In addition, baicalin attenuated inflammatory cell infiltration in the lung of ALI mice and reduced the deposition of complement activation products (C5a, C5b-9) and phosphorylation of NF-κB p65 in lung tissue. CONCLUSION: Baicalin relieves complement alternative pathway activation-induced lung inflammation by inhibition of NF-κB pathway, delaying the progression of ALI.


Subject(s)
Acute Lung Injury , Flavonoids , NF-kappa B , Animals , Flavonoids/pharmacology , Mice , NF-kappa B/metabolism , Acute Lung Injury/drug therapy , Humans , Disease Models, Animal , Male , Complement Pathway, Alternative/drug effects , Pneumonia/drug therapy , Mice, Inbred C57BL , Lung/drug effects , Elapid Venoms/pharmacology
13.
Lung ; 202(5): 513-522, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39259274

ABSTRACT

ALI/ARDS can be a pulmonary manifestation of a systemic inflammatory response or a result of overexpression of the body's normal inflammatory response involving various effector cells, cytokines, and inflammatory mediators, which regulate the body's immune response through different signalling pathways. Forkhead box transcription factors are evolutionarily conserved transcription factors that play a crucial role in various cellular processes, such as cell cycle progression, proliferation, differentiation, migration, metabolism, and DNA damage response. Transcription factors control protein synthesis by regulating gene transcription levels, resulting in diverse biological outcomes. The Fox family plays a role in activating or inhibiting the expression of various molecules related to ALI/ARDS through phosphorylation, acetylation/deacetylation, and control of multiple signalling pathways. An in-depth analysis of the integrated Fox family's role in ALI/ARDS can aid in the development of potential diagnostic and therapeutic targets for the condition.


Subject(s)
Forkhead Transcription Factors , Signal Transduction , Humans , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/drug therapy , Animals , Molecular Targeted Therapy
14.
Eur J Pharmacol ; 982: 176825, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39159715

ABSTRACT

BACKGROUND: Human neutrophil elastase (HNE) is an important contributor to lung diseases such as acute lung injury (ALI) or acute respiratory distress syndrome. Therefore, this study aimed to identify natural HNE inhibitors with anti-inflammatory activity through machine learning algorithms, in vitro assays, molecular dynamic simulation, and an in vivo ALI assay. METHODS: Based on the optimized Discovery Studio two-dimensional molecular descriptors, combined with different molecular fingerprints, six machine learning models were established using the Naïve Bayesian (NB) method to identify HNE inhibitors. Subsequently, the optimal model was utilized to screen 6925 drug-like compounds obtained from the Traditional Chinese Medicine Systems Pharmacy Database and Analysis Platform (TCMSP), followed by ADMET analysis. Finally, 10 compounds with reported anti-inflammatory activity were selected to determine their inhibitory activities against HNE in vitro, and the compounds with the best activity were selected for a 100 ns molecular dynamics simulation and its anti-inflammatory effect was evaluated using Poly (I:C)-induced ALI model. RESULTS: The evaluation of the in vitro HNE inhibition efficiency of the 10 selected compounds showed that the flavonoid tricetin had the strongest inhibitory effect on HNE. The molecular dynamics simulation indicated that the binding of tricetin to HNE was relatively stable throughout the simulation. Importantly, in vivo experiments indicated that tricetin treatment substantially improved the Poly (I:C)-induced ALI. CONCLUSION: The proposed NB model was proved valuable for exploring novel HNE inhibitors, and natural tricetin was screened out as a novel HNE inhibitor, which was confirmed by in vitro and in vivo assays for its inhibitory activities.


Subject(s)
Leukocyte Elastase , Molecular Dynamics Simulation , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Humans , Animals , Male , Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Drug Evaluation, Preclinical , Biological Products/pharmacology , Biological Products/chemistry , Mice , Machine Learning
15.
J Cell Mol Med ; 28(15): e18589, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135202

ABSTRACT

Sepsis causes systemic inflammatory responses and acute lung injury (ALI). Despite modern treatments, sepsis-related ALI mortality remains high. Aqueous extract of Descuraniae Semen (AEDS) exerts anti-endoplasmic reticulum (ER) stress, antioxidant and anti-inflammatory effects. AEDS alleviates inflammation and oedema in ALI. Sodium-potassium-chloride co-transporter isoform 1 (NKCC1) is essential for regulating alveolar fluid and is important in ALI. The NKCC1 activity is regulated by upstream with-no-lysine kinase-4 (WNK4) and STE20/SPS1-related proline/alanine-rich kinase (SPAK). This study aimed to investigate the effects of AEDS on lipopolysaccharide (LPS)-induced ALI model in A549 cells, considering the regulation of ER stress, WNK4-SPAK-NKCC1 cascades, inflammation and apoptosis. Cell viability was investigated by the CCK-8 assay. The expressions of the proteins were assessed by immunoblotting analysis assays. The levels of pro-inflammatory cytokines were determined by ELISA. The expression of cytoplasmic Ca2+ in A549 cells was determined using Fluo-4 AM. AEDS attenuates LPS-induced inflammation, which is associated with increased pro-inflammatory cytokine expression and activation of the WNK4-SPAK-NKCC1 pathway. AEDS inhibits the WNK4-SPAK-NKCC1 pathway by regulating of Bcl-2, IP3R and intracellular Ca2+. WNK4 expression levels are significantly higher in the WNK4-overexpressed transfected A549 cells and significantly decrease after AEDS treatment. AEDS attenuates LPS-induced inflammation by inhibiting the WNK4-SPAK-NKCC1 cascade. Therefore, AEDS is regarded as a potential therapeutic agent for ALI.


Subject(s)
Endoplasmic Reticulum Stress , Inflammation , Lipopolysaccharides , Protein Serine-Threonine Kinases , Signal Transduction , Solute Carrier Family 12, Member 2 , Humans , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , A549 Cells , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Solute Carrier Family 12, Member 2/metabolism , Solute Carrier Family 12, Member 2/genetics , Signal Transduction/drug effects , Apoptosis/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Plant Extracts/pharmacology , Cell Survival/drug effects , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology
16.
Phytomedicine ; 133: 155926, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128302

ABSTRACT

BACKGROUND: Acute lung injury (ALI) is a devastating condition caused by sepsis, pneumonia, trauma, and more recently, COVID-19. SH003, an herbal formula consisted of Astragalus membranaceus, Angelica gigas and Trichosanthes kirilowii, is known for its effects on cancer and immunoregulation. HYPOTHESIS/PURPOSE: Previous studies show SH003 exerts a promising anti-inflammatory effect. This study investigates the effect of modified SH003 on ALI using in silico, in vivo, and in vitro models. STUDY DESIGN AND METHODS: We performed in silico-based analysis of SH003 on ALI-related pathways. C57BL/6 mice were intraperitoneally subjected to lipopolysaccharide (LPS) to induce septic ALI, followed by oral administration of SH003 for 2 weeks. Dexamethasone was used as the positive control. Human peripheral blood-derived polymorphonuclear neutrophils (PMN) were used to investigate the effect and mechanisms of SH003 on neutrophil extracellular trap (NET) formation. RESULTS: Network pharmacology analysis suggested SH003 regulates lung inflammation by modulating NET formation. SH003 significantly reduced mortality in sepsis in vivo by inhibiting local and systemic inflammation, likely via nuclear factor kappa B and mitogen-activated protein kinase pathways-mediated inflammasome suppression. SH003 also decreased NET-related markers in lung tissues and inhibited LPS- and phorbol myristate acetate-induced NET formation in PMN. Cytometry time-of-flight analysis confirmed regulation of NETosis-related pathways by SH003. CONCLUSION: SH003 effectively inhibits excessive immune responses in the lung by suppressing inflammasome activation and NET formation. These findings suggest SH003 as a potential therapeutic agent for septic ALI.


Subject(s)
Acute Lung Injury , Angelica , Astragalus propinquus , Extracellular Traps , Inflammasomes , Lipopolysaccharides , Mice, Inbred C57BL , Neutrophils , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Extracellular Traps/drug effects , Mice , Neutrophils/drug effects , Humans , Inflammasomes/metabolism , Inflammasomes/drug effects , Astragalus propinquus/chemistry , Male , Angelica/chemistry , Drugs, Chinese Herbal/pharmacology , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal
17.
ACS Nano ; 18(32): 21009-21023, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39087239

ABSTRACT

Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.


Subject(s)
Acute Lung Injury , Exosomes , Silicon Dioxide , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Mice , Exosomes/metabolism , Exosomes/chemistry , Silicon Dioxide/chemistry , Verteporfin/pharmacology , Verteporfin/chemistry , Verteporfin/therapeutic use , Nanoparticles/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Male , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Porosity
18.
Int Immunopharmacol ; 141: 112941, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39191119

ABSTRACT

Increasing evidence has demonstrated that coenzyme Q10 (CoQ10) exhibits a range of biological properties. Herein, we explored the protective effect and potential molecular mechanism of CoQ10 on lipopolysaccharide (LPS)-induced acute lung injury (ALI). We found that medium (10 mg/kg) and high (50 mg/kg) doses of CoQ10 ameliorated LPS (50 µg/µL)-induced ALI to varying degrees, as demonstrated by reduced lung coefficient, lower wet/dry weight lung tissue ratio, decreased bronchoalveolar lavage fluid protein concentration, less anatomical and histopathological damage to the lung, and increased expression of proteins related to lung epithelial barrier structure. CoQ10 also alleviated LPS-induced oxidative stress and inflammation mediated by NOD-like receptor protein 3 (NLRP3) by reducing the reactive oxygen species (ROS), malondialdehyde, and mitochondrial ROS concentrations, increasing superoxide dismutase, glutathione, and catalase activity, and decreasing NLRP3 expression at the protein and mRNA levels. Moreover, CoQ10 alleviated structural and functional damage to the mitochondria, inhibited mitochondrial fission, and promoted mitochondrial fusion, mainly by inhibiting phosphorylation of dynamin-related protein 1 (Drp1) at Ser616 and Ser637. Correlation analysis revealed that mitochondrial fission (especially Drp1) was positively correlated with oxidative stress, NLRP3-mediated inflammation, and structural damage to the lung epithelial barrier. Molecular docking analysis showed that CoQ10 binds stably to Drp1, with a binding energy of -5.9 kcal/mol. Furthermore, the use of schaftoside (a Drp1 inhibitor) has further elucidated the mechanism of action of CoQ10. Together, these results suggest that CoQ10 alleviates LPS-induced ALI by regulating mitochondrial dynamics, attenuating oxidative stress, and decreasing NLRP3-medated inflammation, thereby promoting lung epithelial barrier structural remodeling.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Mitochondrial Dynamics , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Ubiquinone , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Animals , Mitochondrial Dynamics/drug effects , Male , Lung/drug effects , Lung/pathology , Lung/metabolism , Lung/immunology , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/chemically induced , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
19.
Int J Pharm ; 664: 124600, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39159858

ABSTRACT

Acute lung injury (ALI) arises from an excessive inflammatory response, usually progressing to acute respiratory distress syndrome (ARDS) if not promptly addressed. There is currently a limited array of effective treatments available for ALI. In this study, we developed disulfide bond-bridged prodrug self-assembled nanoparticles (referred to as DSSS NPs). These nanoparticles were consisted of Dexamethasone (Dex) and stearic acid (SA), and were designed to target and treat ALI. DSSS NPs demonstrated a substantial drug loading capacity with 37.75 % of Dex, which is much higher than conventional nanomedicines (usually < 10 %). Moreover, they exhibited the potential to specifically target injured lung tissue and inflammatory microenvironment-responsive release drugs. Consequently, DSSS NPs reduced significantly the levels of pro-inflammatory cytokines and tissue damage in mice with ALI induced by lipopolysaccharide (LPS). Overall, DSSS NPs offer a promising strategy for treatment of acute lung injury.


Subject(s)
Acute Lung Injury , Anti-Inflammatory Agents , Dexamethasone , Disulfides , Lipopolysaccharides , Nanoparticles , Oxidation-Reduction , Stearic Acids , Dexamethasone/administration & dosage , Dexamethasone/chemistry , Acute Lung Injury/drug therapy , Animals , Nanoparticles/chemistry , Disulfides/chemistry , Mice , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Stearic Acids/chemistry , Male , Drug Liberation , Cytokines/metabolism , Prodrugs/chemistry , Prodrugs/administration & dosage , Humans , Mice, Inbred C57BL , Lung/drug effects , Lung/metabolism , Drug Carriers/chemistry
20.
Eur J Pharmacol ; 982: 176938, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39181224

ABSTRACT

There has been significant global interest in respiratory health driven by the coronavirus disease (COVID-19) and severe environmental pollution. This study explored the potential of schisantherin A (SchA), a compound derived from Schisandra chinensis, to protect against acute pneumoconiosis. We assessed the effects of SchA on phorbol 12-myristate 13-acetate (PMA)-stimulated A549 alveolar epithelial cells and SiO2/TiO2-induced pulmonary injury in mice. In A549 cells, SchA significantly decreased pro-inflammatory mediators such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and interleukin (IL)-8 levels. SchA-mediated reduction in inflammatory mediators was associated with the downregulation of PMA-stimulated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) signaling activation. In SiO2/TiO2-induced lung-injured mice, SchA administration significantly reduced MUC5AC production in lung tissue. SchA administration significantly downregulated the overexpression of NK-κB and the subsequent production of COX-2, iNOS, and NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasomes. It significantly suppressed expected increases in total cell numbers and pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and IL-1ß in the bronchoalveolar lavage fluid (BALF) in SiO2/TiO2-stimulated mice. In contrast, the SiO2/TiO2-mediated decrease in IL-10 levels was significantly improved by SchA treatment. These fundamental results can be used to develop potential treatments involving SchA for acute pneumoconiosis.


Subject(s)
Acute Lung Injury , Cyclooctanes , Nanoparticles , Silicon Dioxide , Titanium , Animals , Silicon Dioxide/toxicity , Titanium/toxicity , Humans , Cyclooctanes/pharmacology , Cyclooctanes/therapeutic use , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , A549 Cells , Male , Nanoparticles/chemistry , Lignans/pharmacology , Lignans/therapeutic use , Mucin 5AC/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Silicosis/pathology , Silicosis/drug therapy , Silicosis/metabolism , Cyclooxygenase 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL