1
|
Master RJ, Karmakar J, Haggie PM, Anthony-Tan J, Chu T, Verkman AS, Anderson MO, Cil O. High potency 3-carboxy-2-methylbenzofuran pendrin inhibitors as novel diuretics. Eur J Med Chem 2025; 283:117133. [PMID: 39642691 DOI: 10.1016/j.ejmech.2024.117133] [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: 09/20/2024] [Revised: 11/07/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Pendrin (SLC26A4) is an anion exchanger expressed in epithelial cells of kidney and lung. Pendrin inhibition is a potential treatment approach for edema, hypertension and inflammatory lung diseases. We have previously identified first-in-class pendrin inhibitors by high-throughput screening, albeit with low potency for pendrin inhibition (IC50 ∼10 μM). Here, we performed a de novo small molecule screen with follow-on structure-activity studies to identify more potent pendrin inhibitors. Screening of 50,000 synthetic small molecules identified four novel classes of pendrin inhibitors with diverse scaffolds, including 5-benzyloxy-2-methylbenzofurans, N-aryl urea substituted 5-methyltryptamines, N-aryl urea substituted anthranilic acids, and substituted N-benzyl 3-carboxyindoles. The most potent inhibitor from the initial screen, a 3-carboxy-2-methylbenzofuran (1a), had IC50 of 4.1 μM. Structure-activity studies using 732 benzofuran analogs identified 1d with IC50 ∼ 0.5 μM for pendrin inhibition. Selectivity studies showed that 1d has minimal or no activity against related ion channels/transporters including SLC26A3, SLC26A6 and CFTR at high concentrations. 1d administration to mice at 10 mg/kg had no effect on urine volume when used alone, but potentiated the diuretic effect of furosemide by 45 %. In conclusion, we have identified novel pendrin inhibitors with greatly improved potency and good in vivo efficacy. These compounds can be used as pharmacological tools to study the roles of pendrin, and potentially developed as drug candidates for edema, hypertension and lung diseases.
Collapse
Affiliation(s)
- Riya J Master
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Joy Karmakar
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Peter M Haggie
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Joseph Anthony-Tan
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Tifany Chu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Alan S Verkman
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Marc O Anderson
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, CA, USA
| | - Onur Cil
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Li H, Wang X, Liang X, Meng M, Zhang H, Li Z, Lin Y, Li J, Ma C. Verapamil inhibits ferroptosis in septic acute lung injury by blocking L-type calcium channels. Biochem Biophys Res Commun 2025; 744:151202. [PMID: 39708394 DOI: 10.1016/j.bbrc.2024.151202] [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: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), result from pulmonary edema and alveolar-capillary barrier disruption due to inflammation, often triggered by conditions like sepsis. Sepsis-induced ALI (SALI) involves extensive damage to vascular endothelium and alveolar epithelium, leading to respiratory failure. Our study explores ferroptosis, an iron-dependent cell death pathway, and calcium dysregulation in SALI. Elevated cytosolic calcium early in ferroptosis exacerbates lipid peroxidation and cellular damage. We investigated verapamil, a calcium channel blocker, and found it reduces calcium influx, alleviates iron overload, and decreases oxidative stress, protecting against ferroptosis-induced apoptosis in lung cells. These insights suggest targeting ferroptosis pathways, including calcium and iron homeostasis, may offer new therapeutic strategies for SALI, potentially improving outcomes in ALI/ARDS.
Collapse
Affiliation(s)
- Hongru Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Xuan Wang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Xiangyang Liang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; School and Hospital of Stomatology, Hebei Medical University, 383 Zhongshan East Road, Shijiazhuang, Hebei, PR China.
| | - Meiqi Meng
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Haixia Zhang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Zixin Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Yushan Lin
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Jihong Li
- Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Cuiqing Ma
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| |
Collapse
|
3
|
Zhang YJ, Chen LY, Lin F, Zhang X, Xiang HF, Rao Q. ROS responsive nanozyme loaded with STING silencing for the treatment of sepsis-induced acute lung injury. Toxicol Appl Pharmacol 2024; 493:117155. [PMID: 39537108 DOI: 10.1016/j.taap.2024.117155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a common complication of sepsis and a leading cause of mortality in septic patients. Studies indicate that STING may play a crucial role in the pathogenesis of sepsis-induced ALI by interacting with the PARP-1/NLRP3 pathway. Therefore, targeting STING inhibition has potential as a novel therapeutic strategy for ALI. However, effective inhibition remains challenging due to the widespread expression of STING across various tissues. In this study, we developed a nanozyme-based drug delivery system, DSPE-TK-mPEG-MnO2@siSTING (abbreviated as DTmM@siSTING), using DSPE-TK-mPEG-MnO2 as the carrier, and characterized it via scanning electron microscopy, dynamic light scattering, nanoparticle size analysis, and gel electrophoresis. To evaluate the therapeutic effects of DTmM@siSTING, an in vitro ALI cell model and an in vivo ALI mouse model were established, assessing the nanozyme's impact on ROS levels, inflammatory responses, and the PARP-1/NLRP3 pathway in sepsis-induced ALI. Results demonstrated that DTmM@siSTING exhibited good physiological stability. In vitro, DTmM@siSTING significantly reduced ROS levels, myeloperoxidase activity, and expression of inflammatory cytokines, while also inhibiting PARP-1/NLRP3 pathway activation. In vivo experiments further revealed that DTmM@siSTING effectively delivered siSTING to the lungs, mitigating sepsis-induced ALI and associated inflammatory responses. Additionally, DTmM@siSTING displayed excellent biocompatibility. In summary, our findings suggest that DTmM@siSTING significantly enhances the therapeutic efficacy of siSTING, alleviating ALI by inhibiting ROS production, inflammatory responses, and activation of the PARP-1/NLRP3 pathway. This novel approach presents a promising therapeutic avenue for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yin-Jin Zhang
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Ling-Yang Chen
- Blood Purification Center, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Feng Lin
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Xia Zhang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China
| | - Hai-Fei Xiang
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| | - Qing Rao
- Department of Anesthesiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, China.
| |
Collapse
|
4
|
Leng T, Zhang L, Ma J, Qu X, Lei B. Intrinsically bioactive multifunctional Poly(citrate-curcumin) for rapid lung injury and MRSA infection therapy. Bioact Mater 2024; 41:158-173. [PMID: 39131630 PMCID: PMC11314446 DOI: 10.1016/j.bioactmat.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Dysregulated inflammation after trauma or infection could result in the further disease and delayed tissue reconstruction. The conventional anti-inflammatory drug treatment suffers to the poor bioavailability and side effects. Herein, we developed an amphiphilic multifunctional poly (citrate-polyglycol-curcumin) (PCGC) nano oligomer with the robust anti-inflammatory activity for treating acute lung injury (ALI) and Methicillin-resistant staphylococcus aureus (MRSA) infected wound. PCGC demonstrated the sustained curcumin release, inherent photoluminescence, good cellular compatibility, hemocompatibility, robust antioxidant activity and enhanced cellular uptake. PCGC could efficiently scavenge nitrogen-based free radicals, oxygen-based free radicals, and intracellular oxygen species, enhance the endothelial cell migration and reduce the expression of pro-inflammatory factors through the NF-κB signal pathway. Combined the anti-inflammation and antioxidant properties, PCGC can shortened the inflammatory process. In animal model of ALI, PCGC was able to reduce the pulmonary edema, bronchial cell infiltration, and lung inflammation, while exhibiting rapid metabolic behavior in vivo. The MRSA-infection wound model showed that PCGC significantly reduced the expression of pro-inflammatory factors, promoted the angiogenesis and accelerated the wound healing. The transcriptome sequencing and molecular mechanism studies further demonstrated that PCGC could inhibit multiple inflammatory related pathways including TNFAIP3, IL-15RA, NF-κB. This work demonstrates that PCGC is efficient in resolving inflammation and promotes the prospect of application in inflammatory diseases as the drug-loaded therapeutic system.
Collapse
Affiliation(s)
- Tongtong Leng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Junping Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoyan Qu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
5
|
Gao N, Liu XY, Chen J, Hu TP, Wang Y, Zhang GQ. Menaquinone-4 Alleviates Sepsis-Associated Acute Lung Injury via Activating SIRT3-p53/SLC7A11 Pathway. J Inflamm Res 2024; 17:7675-7685. [PMID: 39469061 PMCID: PMC11514946 DOI: 10.2147/jir.s486984] [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: 08/24/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
Background Sepsis-associated acute lung injury (SI-ALI) is triggered by various direct or indirect noncardiogenic factors affecting the alveolar epithelium and capillary endothelial cells. Menaquinone-4 (MK-4), a major component of vitamin K, plays a crucial role as an antioxidant by effectively neutralizing reactive oxygen species (ROS) and safeguarding critical biomolecules from oxidative harm within cells. However, the specific mechanisms and clinical implications of MK-4 in SI-ALI are unclear and require further study. Methods Cecal ligation and puncture (CLP) surgery is a commonly used method to induce sepsis in C57BL/6N wild-type mice, and the mice were administered MK-4 at a dosage of 200 mg/kg/day and 3-TYP at 5 mg/kg/day via intraperitoneal injection for 3 days, or erastin (5 mg/kg) 0.5 hours before CLP surgery. The mice were sacrificed 24 hours after CLP surgery, and blood and lung tissue samples were collected. Pathological changes in the lung tissue and oxidative stress levels were detected. The expression levels of Sirt3, acetylated lysine, p53, SLC7A11 ALOX12 and ferroptosis-related proteins were determined. ligation and puncture (CLP). Results In this study, we observed that the lung inflammation was associated with reduced Sirt3 expression and increased acetylated lysine levels. The progression of SI-ALI was mitigated by MK-4 through its role in upregulating Sirt3 expression. MK-4 achieved antioxidant effects by downregulating ROS and inflammatory factor levels. Mechanistically, MK-4 inhibited the p53/SLC7A11 signalling pathway in ferroptosis by inhibiting the acetylation of p53, independent of p53 levels. In addition, MK-4 inhibited ferroptosis independent of GPX4. These findings indicate that MK-4 is a promising novel therapeutic agent for treating SI-ALI and possibly sepsis. Conclusion These experiments revealed that MK-4 acts as a ferroptosis suppressor, increasing the expression of Sirt3, inhibiting the p53/SLC7A11 signalling pathway, and reducing oxidative stress and inflammatory responses, thereby exerting a protective effect against ALI in sepsis.
Collapse
Affiliation(s)
- Nan Gao
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Xiao-Yu Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Jie Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Tian-Peng Hu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Yu Wang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
- Graduate School, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Guo-Qiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| |
Collapse
|
6
|
Liu X, Li Y, Zhang W, Gao N, Chen J, Xiao C, Zhang G. Inhibition of cIAP1/2 reduces RIPK1 phosphorylation in pulmonary endothelial cells and alleviate sepsis-induced lung injury and inflammatory response. Immunol Res 2024; 72:841-850. [PMID: 38748318 DOI: 10.1007/s12026-024-09491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024]
Abstract
Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe complication of sepsis characterized by acute respiratory distress, hypoxemia, and diffuse bilateral pulmonary infiltrates. The regulation of RIPK1 is an important part of the inflammatory response, and cIAP1/2 serves as the E3 ubiquitin ligase for RIPK1. In this study, we investigated the effect and mechanism of cIAP1/2 inhibition on sepsis-induced lung injury. Our results showed that cIAP1/2 inhibition can alleviate sepsis-induced lung injury and reduce the inflammatory response, which is accompanied by downregulation of RIPK1 phosphorylation and ubiquitination. Additionally, cIAP1/2 inhibition led to the up-regulation of programmed cell death, including apoptosis, necroptosis, and pyroptosis, and inhibiting these three cell death pathways can further reduce the inflammatory response, which is similar to the recently discovered programmed cell death pathway PANoptosis. Our findings suggest that cIAP1/2 and PANoptosis inhibition may be a new strategy for treating sepsis-induced lung injury and provide important references for further exploring the mechanism of sepsis-induced lung injury and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Xiaoyu Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Yan Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Weijian Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Peking University, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Nan Gao
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
7
|
Kruk ME, Mehta S, Murray K, Higgins L, Do K, Johnson JE, Wagner R, Wendt CH, O’Connor JB, Harris JK, Laguna TA, Jagtap PD, Griffin TJ. An integrated metaproteomics workflow for studying host-microbe dynamics in bronchoalveolar lavage samples applied to cystic fibrosis disease. mSystems 2024; 9:e0092923. [PMID: 38934598 PMCID: PMC11264604 DOI: 10.1128/msystems.00929-23] [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: 09/01/2023] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Airway microbiota are known to contribute to lung diseases, such as cystic fibrosis (CF), but their contributions to pathogenesis are still unclear. To improve our understanding of host-microbe interactions, we have developed an integrated analytical and bioinformatic mass spectrometry (MS)-based metaproteomics workflow to analyze clinical bronchoalveolar lavage (BAL) samples from people with airway disease. Proteins from BAL cellular pellets were processed and pooled together in groups categorized by disease status (CF vs. non-CF) and bacterial diversity, based on previously performed small subunit rRNA sequencing data. Proteins from each pooled sample group were digested and subjected to liquid chromatography tandem mass spectrometry (MS/MS). MS/MS spectra were matched to human and bacterial peptide sequences leveraging a bioinformatic workflow using a metagenomics-guided protein sequence database and rigorous evaluation. Label-free quantification revealed differentially abundant human peptides from proteins with known roles in CF, like neutrophil elastase and collagenase, and proteins with lesser-known roles in CF, including apolipoproteins. Differentially abundant bacterial peptides were identified from known CF pathogens (e.g., Pseudomonas), as well as other taxa with potentially novel roles in CF. We used this host-microbe peptide panel for targeted parallel-reaction monitoring validation, demonstrating for the first time an MS-based assay effective for quantifying host-microbe protein dynamics within BAL cells from individual CF patients. Our integrated bioinformatic and analytical workflow combining discovery, verification, and validation should prove useful for diverse studies to characterize microbial contributors in airway diseases. Furthermore, we describe a promising preliminary panel of differentially abundant microbe and host peptide sequences for further study as potential markers of host-microbe relationships in CF disease pathogenesis.IMPORTANCEIdentifying microbial pathogenic contributors and dysregulated human responses in airway disease, such as CF, is critical to understanding disease progression and developing more effective treatments. To this end, characterizing the proteins expressed from bacterial microbes and human host cells during disease progression can provide valuable new insights. We describe here a new method to confidently detect and monitor abundance changes of both microbe and host proteins from challenging BAL samples commonly collected from CF patients. Our method uses both state-of-the art mass spectrometry-based instrumentation to detect proteins present in these samples and customized bioinformatic software tools to analyze the data and characterize detected proteins and their association with CF. We demonstrate the use of this method to characterize microbe and host proteins from individual BAL samples, paving the way for a new approach to understand molecular contributors to CF and other diseases of the airway.
Collapse
Affiliation(s)
- Monica E. Kruk
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| | - Subina Mehta
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| | - Kevin Murray
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
- Center for Metabolomics and Proteomics, University of Minnesota, Minneapolis, Minnesota, USA
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
- Center for Metabolomics and Proteomics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katherine Do
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| | - James E. Johnson
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Reid Wagner
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chris H. Wendt
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
- Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - John B. O’Connor
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
| | - J. Kirk Harris
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Theresa A. Laguna
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Pratik D. Jagtap
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| | - Timothy J. Griffin
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minneapolis, Minnesota, USA
| |
Collapse
|
8
|
Lu L, Zhu C, Xu J, Hu Y, Dai J, Wang S, Wei T. Therapeutic Effects of Lifei Decoction in a Murine Model of COPD Induced by LPS and Cigarette Smoke. Int J Chron Obstruct Pulmon Dis 2024; 19:957-967. [PMID: 38650680 PMCID: PMC11034514 DOI: 10.2147/copd.s449521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction The Lifei Decoction (LD) is a commonly utilized Chinese medicine for the treatment of sepsis and bronchial inflammation. However, its therapeutic potential in chronic obstructive pulmonary disease (COPD) remains unknown. Therefore, the objective of this study was to investigate the therapeutic efficacy and underlying mechanism of LD in a mouse model of COPD induced by cigarette smoke (CS) combined with lipopolysaccharide (LPS). Methods Hematoxylin-eosin (H&E) staining was employed to observe the pathological alterations in lung tissue, while ELISA was utilized for the detection of levels of inflammatory factors in both lung tissue and bronchoalveolar lavage fluid (BALF). Additionally, Western blot analysis was conducted to assess the expression of p-NF-κB, GDF11, ZO-1, and Occludin-1 proteins. The changes in intestinal flora were evaluated using the viable bacteria count method. Results The administration of LD demonstrates significant efficacy in mitigating pulmonary tissue damage in a murine model, while concurrently inhibiting the activation of the inflammatory pathway NF-κB to attenuate the levels of pro-inflammatory factors. Moreover, LD exhibits the capacity to enhance the expression of intestinal functional proteins ZO-1 and Occludin-1, thereby rectifying dysbiosis within the gut microbiota. Conclusion The LD shows great promise as a potential treatment for COPD.
Collapse
Affiliation(s)
- Liguo Lu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, People’s Republic of China
| | - Chengdong Zhu
- Department of Traditional Chinese Medicine, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, People’s Republic of China
| | - Jian Xu
- Taian Maternal and Child Health Hospital, Tai An, Shandong, People’s Republic of China
| | - Yulan Hu
- Department of Respiratory and Critical Care Medicine, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, People’s Republic of China
| | - Juxiang Dai
- Department of Respiratory and Critical Care Medicine, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, People’s Republic of China
| | - Sheng Wang
- Department of Respiratory and Critical Care Medicine, Shuyang Hospital, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian, People’s Republic of China
| | - Tao Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, People’s Republic of China
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
9
|
Lee D, Hong JH. Chloride/Multiple Anion Exchanger SLC26A Family: Systemic Roles of SLC26A4 in Various Organs. Int J Mol Sci 2024; 25:4190. [PMID: 38673775 PMCID: PMC11050216 DOI: 10.3390/ijms25084190] [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: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Solute carrier family 26 member 4 (SLC26A4) is a member of the SLC26A transporter family and is expressed in various tissues, including the airway epithelium, kidney, thyroid, and tumors. It transports various ions, including bicarbonate, chloride, iodine, and oxalate. As a multiple-ion transporter, SLC26A4 is involved in the maintenance of hearing function, renal function, blood pressure, and hormone and pH regulation. In this review, we have summarized the various functions of SLC26A4 in multiple tissues and organs. Moreover, the relationships between SLC26A4 and other channels, such as cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and sodium chloride cotransporter, are highlighted. Although the modulation of SLC26A4 is critical for recovery from malfunctions of various organs, development of specific inducers or agonists of SLC26A4 remains challenging. This review contributes to providing a better understanding of the role of SLC26A4 and development of therapeutic approaches for the SLC26A4-associated hearing loss and SLC26A4-related dysfunction of various organs.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, GAIHST (Gachon Advanced Institute for Health Sciences and Technology), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
10
|
Wu Q, Liu C, Liu D, Wang Y, Qi H, Liu X, Zhang Y, Chen H, Zeng Y, Li J. Polystyrene nanoplastics-induced lung apoptosis and ferroptosis via ROS-dependent endoplasmic reticulum stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169260. [PMID: 38086481 DOI: 10.1016/j.scitotenv.2023.169260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
It has been shown that exposure to nanoplastics (MNPs) through inhalation can induce pulmonary toxicity, but the toxicological mechanism of MNPs on the respiratory system remains unclear. Therefore, we explored the toxicological mechanism of exposure to polystyrene nanoplastics (PS-NPs) (0.05, 0.15, 0.2 mg/mL) on BEAS-2B cells. Results revealed that PS-NPs induce oxidative stress, increased apoptosis rate measured by flow cytometry, the key ferroptosis protein (GPX4 and FTH1) reduction, increased iron content, mitochondrial alterations, and increased malondialdehyde (MDA) level. Besides, consistent results were observed in mice exposed to PS-NPs (5 mg/kg/2d, 10 mg/kg/2d). Thus, we proved that PS-NPs induced cell death and lung damage through apoptosis and ferroptosis. In terms of mechanism, the elevation of the endoplasmic reticulum (ER) stress protein expression (IRE1α, PERK, XBP1S, and CHOP) revealed that PS-NPs induce lung damage by activating the two main ER stress pathways. Furthermore, the toxicological effects of PS-NPs observed in this study are attenuated by the ROS inhibitor N-acetylcysteine (NAC). Collectively, NPs-induced apoptosis and ferroptosis are attenuated by NAC via inhibiting the ROS-dependent ER stress in vitro and in vivo. This improves our understanding of the mechanism by which PS-NPs exposure leads to pulmonary injury and the potential protective effects of NAC.
Collapse
Affiliation(s)
- Qiumei Wu
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chao Liu
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Dan Liu
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yougang Wang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Haomin Qi
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xudong Liu
- Department of Brewing Engineering, Moutai Institute, Renhuai 564507, China
| | - Yuchao Zhang
- Department of Brewing Engineering, Moutai Institute, Renhuai 564507, China
| | - Haiyu Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jinquan Li
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
11
|
Wang L, Hoang A, Gil-Iturbe E, Laganowsky A, Quick M, Zhou M. Mechanism of anion exchange and small-molecule inhibition of pendrin. Nat Commun 2024; 15:346. [PMID: 38184688 PMCID: PMC10771415 DOI: 10.1038/s41467-023-44612-1] [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: 06/24/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Pendrin (SLC26A4) is an anion exchanger that mediates bicarbonate (HCO3-) exchange for chloride (Cl-) and is crucial for maintaining pH and salt homeostasis in the kidney, lung, and cochlea. Pendrin also exports iodide (I-) in the thyroid gland. Pendrin mutations in humans lead to Pendred syndrome, causing hearing loss and goiter. Inhibition of pendrin is a validated approach for attenuating airway hyperresponsiveness in asthma and for treating hypertension. However, the mechanism of anion exchange and its inhibition by drugs remains poorly understood. We applied cryo-electron microscopy to determine structures of pendrin from Sus scrofa in the presence of either Cl-, I-, HCO3- or in the apo-state. The structures reveal two anion-binding sites in each protomer, and functional analyses show both sites are involved in anion exchange. The structures also show interactions between the Sulfate Transporter and Anti-Sigma factor antagonist (STAS) and transmembrane domains, and mutational studies suggest a regulatory role. We also determine the structure of pendrin in a complex with niflumic acid (NFA), which uncovers a mechanism of inhibition by competing with anion binding and impeding the structural changes necessary for anion exchange. These results reveal directions for understanding the mechanisms of anion selectivity and exchange and their regulations by the STAS domain. This work also establishes a foundation for analyzing the pathophysiology of mutations associated with Pendred syndrome.
Collapse
Affiliation(s)
- Lie Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Anthony Hoang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, TX, USA
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Area Neuroscience - Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
| | - Ming Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Geertsma ER, Oliver D. SLC26 Anion Transporters. Handb Exp Pharmacol 2024; 283:319-360. [PMID: 37947907 DOI: 10.1007/164_2023_698] [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] [Indexed: 11/12/2023]
Abstract
Solute carrier family 26 (SLC26) is a family of functionally diverse anion transporters found in all kingdoms of life. Anions transported by SLC26 proteins include chloride, bicarbonate, and sulfate, but also small organic dicarboxylates such as fumarate and oxalate. The human genome encodes ten functional homologs, several of which are causally associated with severe human diseases, highlighting their physiological importance. Here, we review novel insights into the structure and function of SLC26 proteins and summarize the physiological relevance of human members.
Collapse
Affiliation(s)
- Eric R Geertsma
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Dominik Oliver
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University Marburg, Marburg, Germany.
- Center for Mind, Brain and Behavior (CMBB), Universities of Marburg and Giessen, Marburg, Giessen, Germany.
| |
Collapse
|
13
|
Liu X, Chen J, Li Z, Gao N, Zhang G. CIAP1/2 can regulate the inflammatory response and lung injury induced by apoptosis in septic rats. J Investig Med 2024; 72:100-111. [PMID: 37784217 DOI: 10.1177/10815589231207102] [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] [Indexed: 10/04/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), induced by sepsis, is predominantly caused by inflammation injury. However, there is no clear consensus on how to regulate the inflammatory response. The TNF pathway is one of the primary inflammatory pathways activated in sepsis. cIAP1/2, an essential E3 ubiquitin ligase in the TNF pathway, plays a pivotal role in positively regulating the activation of nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways to promote inflammation while inhibiting apoptosis. We found that Birc2 is the only differential expression gene in TNF pathway, and both cIAP1/2 upregulated in lung lysate with worsen lung injury. However, upon inhibiting cIAP1/2 using AZD5582, lung cell apoptosis was reactivated, and a significant improvement in lung injury was observed. Our study shows that cIAP1/2 expression increased in the lung tissue of a CLP rat ALI model. Inhibiting cIAP1/2 with AZD5582, a second mitochondria-derived activator of caspases (SMAC) mimetic, induced increased apoptosis and reduced lung injury. Therefore, inhibiting cIAP1/2 can alleviate sepsis-induced ALI, providing a new target for regulating organ damage induced by sepsis-induced inflammatory responses.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Chen
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhonghao Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Nan Gao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
14
|
Zajac M, Lepissier A, Dréano E, Chevalier B, Hatton A, Kelly-Aubert M, Guidone D, Planelles G, Edelman A, Girodon E, Hinzpeter A, Crambert G, Pranke I, Galietta LJV, Sermet-Gaudelus I. Putting bicarbonate on the spot: pharmacological insights for CFTR correction in the airway epithelium. Front Pharmacol 2023; 14:1293578. [PMID: 38149052 PMCID: PMC10750368 DOI: 10.3389/fphar.2023.1293578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction: Cystic fibrosis (CF) is caused by defective Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) proteins. CFTR controls chloride (Cl-) and bicarbonate (HCO3 -) transport into the Airway Surface Liquid (ASL). We investigated the impact of F508del-CFTR correction on HCO3 - secretion by studying transepithelial HCO3 - fluxes. Methods: HCO3 - secretion was measured by pH-stat technique in primary human respiratory epithelial cells from healthy subjects (WT) and people with CF (pwCF) carrying at least one F508del variant. Its changes after CFTR modulation by the triple combination VX445/661/770 and in the context of TNF-α+IL-17 induced inflammation were correlated to ASL pH and transcriptional levels of CFTR and other HCO3 - transporters of airway epithelia such as SLC26A4 (Pendrin), SLC26A9 and NBCe1. Results: CFTR-mediated HCO3 - secretion was not detected in F508del primary human respiratory epithelial cells. It was rescued up to ∼ 80% of the WT level by VX-445/661/770. In contrast, TNF-α+IL-17 normalized transepithelial HCO3 - transport and increased ASL pH. This was related to an increase in SLC26A4 and CFTR transcript levels. VX-445/661/770 induced an increase in pH only in the context of inflammation. Effects on HCO3 - transport were not different between F508del homozygous and F508del compound heterozygous CF airway epithelia. Conclusion: Our studies show that correction of F508del-CFTR HCO3 - is not sufficient to buffer acidic ASL and inflammation is a key regulator of HCO3 - secretion in CF airways. Prediction of the response to CFTR modulators by theratyping should take into account airway inflammation.
Collapse
Affiliation(s)
- Miroslaw Zajac
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Agathe Lepissier
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Elise Dréano
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Benoit Chevalier
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Aurélie Hatton
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Mairead Kelly-Aubert
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | - Aleksander Edelman
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Emmanuelle Girodon
- Université de Paris-Cité, Paris, France
- Service de Médecine Génomique des Maladies de Système et d’Organe, Hôpital Cochin, Paris, France
| | - Alexandre Hinzpeter
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Gilles Crambert
- U1138/CNRS ERL 8228, Centre de Recherche des Cordeliers, Paris, France
| | - Iwona Pranke
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Isabelle Sermet-Gaudelus
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Université de Paris-Cité, Paris, France
- Centre de Référence Maladie Rare Pour La Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- European Reference Network for Rare Diseases, Frankfurt, Belgium
| |
Collapse
|
15
|
Mucke HA. Patent highlights December 2022-January 2023. Pharm Pat Anal 2023; 12:151-158. [PMID: 37801039 DOI: 10.4155/ppa-2023-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
|
16
|
Ma A, Feng Z, Li Y, Wu Q, Xiong H, Dong M, Cheng J, Wang Z, Yang J, Kang Y. Ferroptosis-related signature and immune infiltration characterization in acute lung injury/acute respiratory distress syndrome. Respir Res 2023; 24:154. [PMID: 37301835 DOI: 10.1186/s12931-023-02429-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/19/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one of the most life-threatening diseases in the intensive care unit with high mortality and morbidity. Ferroptosis is a newly discovered immune related cell death that is associated with various lung diseases. However, the role of immune-mediated ferroptosis in ALI/ARDS has not been elucidated. METHOD We analyzed two Gene Expression Omnibus (GEO) datasets (GSE2411 and GSE109913) and extracted characteristic ferroptosis-related genes (FRGs) between the control and ALI groups through bioinformatic analysis. Then, we prospectively collected bronchoalveolar lavage fluid (BALF) from patients with ARDS and verified the expression of characteristic FRGs. Lastly, we constructed the ALI/ARDS model induced by LPS and isolated the primary neutrophils of mice. Erastin, an ferroptosis inducer, was used at the cellular level to verify the effect of neutrophils on ferroptosis in lung epithelium cells. RESULT We identified three characteristic FRGs, Cp, Slc39a14 and Slc7a11, by analyzing two gene expression profiling datasets. Immune infiltration analysis showed that the three characteristic genes were significantly positively correlated with the infiltration levels of neutrophils. We collected BALF from 59 ARDS patients to verify the expression of Cp, Slc7a11 and Slc39a14 in humans. The results showed that Cp was elevated in patients with severe ARDS (p = 0.019), Slc7a11 was significantly elevated in patients with moderate ARDS (p = 0.021) relative to patients with mild ARDS. The levels of neutrophils in the peripheral blood of ARDS patients were positively correlated with the expression levels of Slc7a11 (Pearson's R2 = 0.086, p = 0.033). Three characteristic FRGs were significantly activated after the onset of ferroptosis (6 h) early in LPS induced ALI model, and that ferroptosis was alleviated after the organism compensated within 12 to 48 h. We extracted primary activated neutrophils from mice and co-cultured them with MLE-12 in transwell, Slc7a11, Cp and Slc39a14 in MLE-12 cells were significantly upregulated as the number of neutrophils increased. The results showed that neutrophil infiltration alleviated erastin-induced MDA accumulation, GSH depletion, and divalent iron accumulation, accompanied by upregulation of Slc7a11 and Gpx4, implying the existence of a compensatory effect of lipid oxidation in neutrophils after acute lung injury in the organism. CONCLUSION We identified three immune-mediated ferroptosis genes, namely, Cp, Slc7a11 and Slc39a14, which possibly regulated by neutrophils during the development of ALI, and their pathways may be involved in anti-oxidative stress and anti-lipid metabolism. Thus, the present study contributes to the understanding of ALI/ARDS and provide novel targets for future immunotherapeutic.
Collapse
Affiliation(s)
- Aijia Ma
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zhongxue Feng
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yang Li
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Qin Wu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Huaiyu Xiong
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Meiling Dong
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Jiangli Cheng
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Zhenling Wang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, No. 37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
17
|
Chen H, Dong J, Yang L, Song Y, Zhu H, Yang Y. Mitochondrial targeted fluorescent nitrite peroxide probe for dynamic monitoring of cellular lung injury. Anal Biochem 2023; 668:115114. [PMID: 36907310 DOI: 10.1016/j.ab.2023.115114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/26/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
Herein, a mitochondrial targeted fluorescent nitrite peroxide probe CHP for dynamic monitoring of cellular lung injury was developed. For the practical delivery and selectivity, the structural features including pyridine head and borate recognition group were selected. CHP could respond to ONOO- with the 585 nm fluorescence signal. The detecting system indicated advantages such as wide linear range (0.0-30 μM), high sensitivity (LOD = 0.18 μM), high selectivity and steadiness under different environmental conditions including pH (3.0-10.0), time (48 h) and medium. In living A549 cells, the response of CHP towards ONOO- showed dose-dependent and time-dependent tendencies. The co-localization suggested that CHP could achieve mitochondrial targeting. Moreover, CHP could monitor the variation of endogenous ONOO- level and the cellular lung injury induced by LPS.
Collapse
Affiliation(s)
- Hao Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Junming Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Longyang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Yongchun Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China.
| | - Hailiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China.
| | - Yushun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China; Jinhua Advanced Research Institute, Jinhua, 321019, China.
| |
Collapse
|
18
|
Tu Y, Liu J, Kong D, Guo X, Li J, Long Z, Peng J, Wang Z, Wu H, Liu P, Liu R, Yu W, Li W. Irisin drives macrophage anti-inflammatory differentiation via JAK2-STAT6-dependent activation of PPARγ and Nrf2 signaling. Free Radic Biol Med 2023; 201:98-110. [PMID: 36940733 DOI: 10.1016/j.freeradbiomed.2023.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023]
Abstract
Irisin is an exercise-induced myokine that alleviates inflammation and obesity. The induction of anti-inflammatory (M2) macrophage is facilitated for treatment of sepsis and associated lung damage. However, whether irisin drives macrophage M2 polarization remains unclear. Here, we found that irisin induced-macrophage anti-inflammatory differentiation in vivo using an LPS-induced septic mice model and in vitro using RAW64.7 cells and bone marrow-derived macrophages (BMDMs). Irisin also promoted the expression, phosphorylation, and nuclear translocation of peroxisome proliferator-activated receptor gamma (PPAR-γ) and nuclear factor-erythroid 2-related factor 2 (Nrf2). Inhibition or knockdown of PPAR-γ and Nrf2 abolished irisin-induced accumulation of M2 macrophage markers, such as interleukin (IL)-10 and Arginase 1. Furthermore, dual-luciferase reporter and chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR) assays confirmed that STAT6 boosts PPAR-γ and Nrf2 transcription by binding to their DNA promoters in irisin-stimulated macrophages. In contrast, STAT6 shRNA blocked the irisin-induced activation of Pparγ, Nrf2, and related downstream genes. Moreover, the interaction of irisin with its ligand integrin αVβ5 remarkably promoted Janus kinase 2 (JAK2) phosphorylation, while inhibition or knockdown of integrin αVβ5 and JAK2 attenuated the activation of STAT6, PPAR-γ, and Nrf2 signaling. Interestingly, co-immunoprecipitation (Co-IP) assay also revealed that the binding between JAK2 and integrin αVβ5 is critical for irisin-induced macrophage anti-inflammatory differentiation by enhancing the activation of the JAK2-STAT6 pathway. In conclusion, irisin boosted M2 macrophage differentiation by inducing JAK2-STAT6-dependent transcriptional activation of the PPAR-γ-related anti-inflammatory system and Nrf2-related antioxidant genes. The findings of this study suggest that the administration of irisin is a novel and promising therapeutic strategy for infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yongmei Tu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China; School of Public Health, Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, China
| | - Jiangzheng Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Deqin Kong
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaojie Guo
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiawei Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zi Long
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Peng
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhao Wang
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Wu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Penghui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Liu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Weihua Yu
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Wenli Li
- Department of Toxicology, Shaanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
19
|
Elucidation of the underlying mechanism of Hua-ban decoction in alleviating acute lung injury by an integrative approach of network pharmacology and experimental verification. Mol Immunol 2023; 156:85-97. [PMID: 36913767 DOI: 10.1016/j.molimm.2023.02.013] [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: 08/01/2022] [Revised: 12/29/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
The pathogenic hyper-inflammatory response has been regarded as the major cause of the severity and death related to acute lung injury (ALI). Hua-ban decoction (HBD) is a classical prescription in traditional Chinese medicine (TCM). It has been extensively used to treat inflammatory diseases; however, its bioactive components and therapeutic mechanisms remain unclear. Here, we established a lipopolysaccharide (LPS)-induced ALI model that presents a hyperinflammatory process to explore the pharmaco-dynamic effect and underlying molecular mechanism of HBD on ALI. In vivo, we confirmed that in LPS-induced ALI mice, HBD improved pulmonary injury by via down-regulating the expression of proinflammatory cytokines, including IL-6, TNF-α, and macrophage infiltration, as well as macrophage M1 polarization. Moreover, in vitro experiments in LPS-stimulated macrophages demonstrated that the potential bioactive compounds of HBD inhibited the secretion of IL-6 and TNF-α. Mechanically, the data revealed that HBD treatment of LPS-induced ALI acted via NF-κB pathway, which regulated macrophage M1 polarization. Additionally, two major HBD compounds, i.e., quercetin and kaempferol, showed a high binding affinity with p65 and IkBα. In conclusion, the data obtained in this study demonstrated the therapeutic effects of HBD, which indicates the possibility for the development of HBD as a potential treatment for ALI.
Collapse
|
20
|
Chantree P, Tarasuk M, Prathaphan P, Ruangtong J, Jamklang M, Chumkiew S, Martviset P. Type I Cystatin Derived from Fasciola gigantica Suppresses Macrophage-Mediated Inflammatory Responses. Pathogens 2023; 12:pathogens12030395. [PMID: 36986318 PMCID: PMC10051455 DOI: 10.3390/pathogens12030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/05/2023] Open
Abstract
There is an inverse relationship between the high incidence of helminth infection and the low incidence of inflammatory disease. Hence, it may be that helminth molecules have anti-inflammatory effects. Helminth cystatins are being extensively studied for anti-inflammatory potential. Therefore, in this study, the recombinant type I cystatin (stefin-1) of Fasciola gigantica (rFgCyst) was verified to have LPS-activated anti-inflammatory potential, including in human THP-1-derived macrophages and RAW 264.7 murine macrophages. The results from the MTT assay suggest that rFgCyst did not alter cell viability; moreover, it exerted anti-inflammatory activity by decreasing the production of proinflammatory cytokines and mediators, including IL-1β, IL-6, IL-8, TNF-α, iNOS, and COX-2 at the gene transcription and protein expression levels, as determined by qRT-PCR and Western blot analysis, respectively. Further, the secretion levels of IL-1β, IL-6, and TNF-α determined by ELISA and the NO production level determined by the Griess test were decreased. Furthermore, in Western blot analysis, the anti-inflammatory effects involved the downregulation of pIKKα/β, pIκBα, and pNF-κB in the NF-κB signaling pathway, hence reducing the translocation from the cytosol into the nucleus of pNF-κB, which subsequently turned on the gene of proinflammatory molecules. Therefore, cystatin type 1 of F. gigantica is a potential candidate for inflammatory disease treatment.
Collapse
Affiliation(s)
- Pathanin Chantree
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Mayuri Tarasuk
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Parisa Prathaphan
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Jittiporn Ruangtong
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Mantana Jamklang
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Sirilak Chumkiew
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pongsakorn Martviset
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Correspondence: ; Tel.: +66-863590511
| |
Collapse
|
21
|
Chantree P, Martviset P, Thongsepee N, Sangpairoj K, Sornchuer P. Anti-Inflammatory Effect of Garcinol Extracted from Garcinia dulcis via Modulating NF-κB Signaling Pathway. Nutrients 2023; 15:nu15030575. [PMID: 36771283 PMCID: PMC9918937 DOI: 10.3390/nu15030575] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/14/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Garcinia is a significant medicinal plant with many beneficial phytoconstituents, including garcinol. This study investigated the anti-inflammatory effect of garcinol isolated from Garcinia dulcis fruit in LPS-activated THP-1 and Raw 264.7 macrophages. The results demonstrated that the low concentration of garcinol did not alter cell viability. Furthermore, co-incubation of garcinol with LPS inhibited the production of pro-inflammatory cytokines, including TNF-α, IL-8, IL-6, IL-1β, and pro-inflammatory mediators, including iNOS and COX-2 at the mRNA and protein expression levels. Garcinol also decreased the secretion of TNF-α, IL-6, IL-1β, PGE2, and NO. Moreover, the anti-inflammatory effects involved an alteration in the NF-κB signaling pathway. Downregulation of pIKKα/β, pIκBα, and pNF-κB was observed, hence reducing the translocation of pNF-κB from the cytosol into the nucleus, which subsequently decreased the production of pro-inflammatory molecules. Therefore, garcinol isolated from Garcinia dulcis is a potential candidate as an anti-inflammatory agent for inflammation-related disease treatment.
Collapse
Affiliation(s)
- Pathanin Chantree
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Correspondence: ; Tel.: +66-846-171-817
| | - Pongsakorn Martviset
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Nattaya Thongsepee
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Kant Sangpairoj
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Phornphan Sornchuer
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| |
Collapse
|
22
|
Fan XP, Huang J, Ren TB, Yuan L, Zhang XB. De Novo Design of Activatable Photoacoustic/Fluorescent Probes for Imaging Acute Lung Injury In Vivo. Anal Chem 2023; 95:1566-1573. [PMID: 36584357 DOI: 10.1021/acs.analchem.2c04642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Effective monitoring of the physiological progression of acute lung injury (ALI) in real time is crucial for early theranostics to reduce its high mortality. In particular, activatable fluorescence and photoacoustic molecule probes have attracted attention to assess ALI by detecting related indicators. However, the existing fluorophores often encounter issues of low retention in the lungs and slow clearance from the body, which compromise the probe's actual capability for in situ imaging by intravenous injection in vivo. Herein, a novel near-infrared hemicyanines fluorophore (FJH) bearing a quaternary ammonium group was first developed by combining with the rational design and screening strategy. The properties of good hydrophilicity and blood circulation effectively enable FJH accumulation for lung imaging. Inspired by the high retention efficiency, the probe FJH-C that turns on fluorescence and photoacoustic signals in response to the ALI indicator (esterase) was subsequently synthesized. Notably, the probe FJH-C successfully achieved the selectivity and sensitivity toward esterase in vitro and in living cells. More importantly, FJH-C can be further used to assess lipopolysaccharides and silica-induced ALI through the desired fluo-photoacoustic signal. Therefore, this study not only shows the first activatable probe for real-time imaging of lung function but also highlights the fluorophore structure with high lung retention. It is believed that FJH and FJH-C can serve as an efficient platform to reveal the pathological progression of other lung diseases for early diagnosis and medical intervention.
Collapse
Affiliation(s)
- Xiao-Peng Fan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Hebei University, Baoding 071002, P.R. China
| | - Jing Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tian-Bing Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
23
|
Zhang X, Wu D, Tian Y, Chen X, Lan J, Wei F, Li Y, Luo Y, Sun X. Ganoderma lucidum polysaccharides ameliorate lipopolysaccharide-induced acute pneumonia via inhibiting NRP1-mediated inflammation. PHARMACEUTICAL BIOLOGY 2022; 60:2201-2209. [PMID: 36373992 PMCID: PMC9665083 DOI: 10.1080/13880209.2022.2142615] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Ganoderma lucidum polysaccharides (GLP), from Ganoderma lucidum (Leyss. ex Fr.) Karst. (Ganodermataceae), are reported to have anti-inflammatory effects, including anti-neuroinflammation and anti-colitis. Nevertheless, the role of GLP in acute pneumonia is unknown. OBJECTIVE To explore the protective role of GLP against LPS-induced acute pneumonia and investigate possible mechanisms. MATERIALS AND METHODS GLP were extracted and used for high-performance liquid chromatography (HPLC) analysis after acid hydrolysis and PMP derivatization. Sixty C57BL/6N male mice were randomly divided into six groups: Sham, Model, LPS + GLP (25, 50 and 100 mg/kg/d administered intragastrically for two weeks) and LPS + dexamethasone (6 mg/kg/d injected intraperitoneally for one week). Acute pneumonia mouse models were established by intratracheal injection of LPS. Haematoxylin and eosin (H&E) staining was examined to evaluate lung lesions. ELISA and quantitative real-time PCR were employed to assess inflammatory factors expression. Western blots were carried out to measure Neuropilin-1 expression and proteins related to apoptosis and autophagy. RESULTS GLP suppressed inflammatory cell infiltration. In BALF, cell counts were 1.1 × 106 (model) and 7.1 × 105 (100 mg/kg). Release of GM-CSF and IL-6 was reduced with GLP (25, 50 and 100 mg/kg) treatment. The expression of genes IL-1β, IL-6, TNF-α and Saa3 was reduced. GLP treatment also suppressed the activation of Neuropilin-1 (NRP1), upregulated the levels of Bcl2/Bax and LC3 and led to downregulation of the ratio C-Caspase 3/Caspase 3 and P62 expression. DISCUSSION AND CONCLUSIONS GLP could protect against LPS-induced acute pneumonia through multiple mechanisms: blocking the infiltration of inflammatory cells, inhibiting cytokine secretion, suppressing NRP1 activation and regulating pneumonocyte apoptosis and autophagy.
Collapse
Affiliation(s)
- Xuelian Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Daoshun Wu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Xiangdong Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jin Lan
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wei
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ye Li
- Ganoherb (Fujian) Technology Corporation, Nanping, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Guidone D, Buccirossi M, Scudieri P, Genovese M, Sarnataro S, De Cegli R, Cresta F, Terlizzi V, Planelles G, Crambert G, Sermet I, Galietta LJ. Airway surface hyperviscosity and defective mucociliary transport by IL-17/TNF-α are corrected by β-adrenergic stimulus. JCI Insight 2022; 7:164944. [PMID: 36219481 DOI: 10.1172/jci.insight.164944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/05/2022] [Indexed: 12/15/2022] Open
Abstract
The fluid covering the surface of airway epithelia represents a first barrier against pathogens. The chemical and physical properties of the airway surface fluid are controlled by the activity of ion channels and transporters. In cystic fibrosis (CF), loss of CFTR chloride channel function causes airway surface dehydration, bacterial infection, and inflammation. We investigated the effects of IL-17A plus TNF-α, 2 cytokines with relevant roles in CF and other chronic lung diseases. Transcriptome analysis revealed a profound change with upregulation of several genes involved in ion transport, antibacterial defense, and neutrophil recruitment. At the functional level, bronchial epithelia treated in vitro with the cytokine combination showed upregulation of ENaC channel, ATP12A proton pump, ADRB2 β-adrenergic receptor, and SLC26A4 anion exchanger. The overall result of IL-17A/TNF-α treatment was hyperviscosity of the airway surface, as demonstrated by fluorescence recovery after photobleaching (FRAP) experiments. Importantly, stimulation with a β-adrenergic agonist switched airway surface to a low-viscosity state in non-CF but not in CF epithelia. Our study suggests that CF lung disease is sustained by a vicious cycle in which epithelia cannot exit from the hyperviscous state, thus perpetuating the proinflammatory airway surface condition.
Collapse
Affiliation(s)
- Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Paolo Scudieri
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy
| | - Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Sergio Sarnataro
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Federico Cresta
- Centro Fibrosi Cistica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Vito Terlizzi
- Meyer Children's Hospital, Cystic Fibrosis Regional Reference Center, Department of Paediatric Medicine, Firenze, Italy
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France.,CNRS EMR 8228, Paris, France
| | - Gilles Crambert
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France.,CNRS EMR 8228, Paris, France
| | | | - Luis Jv Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Translational Medical Sciences (DISMET), University of Napoli "Federico II", Napoli, Italy
| |
Collapse
|
25
|
Peng SW, Sheng JM, Feng BS, Peng KP, Tian GX, Liang CB, Liu MH, Xie HQ, Shu Q, Li Y, Yang PC. Identification of mite-specific eosinophils in the colon of patients with ulcerative colitis. Autoimmunity 2022; 55:549-558. [PMID: 36062759 DOI: 10.1080/08916934.2022.2114467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The pathogenesis of ulcerative colitis (UC) is unclear. House dust mite (HDM) is associated with immune inflammation in the body. This study is designed to identify the association between HDM and UC clinical symptoms. UC patients (n = 86) and non-UC control (NC) subjects (n = 64) were recruited. Colon lavage fluids (CLF) were collected from HDM skin prick test positive patients during colonoscopy, and analyzed by immunological approaches. HDM was detected in fecal samples, which was positively correlated with UC clinical symptoms. HDM-specific eosinophils and Th2 cells were detected in CLF, which could be specifically activated by exposing to HDM in the culture. Direct exposure to HDM induced eosinophil activation in the colon of UC patients. UC patients displayed elevated levels of Th2 cytokines in the serum. UC clinical symptom scores were positively correlated with serum levels of Th2 cytokines. HDM was detected in UC patients' stools, which was positively correlated with UC clinical symptoms. Direct exposure to HDM could trigger eosinophilic activation of the colon.
Collapse
Affiliation(s)
- Shu-Wang Peng
- Department of Gastrointestinal and Thyroid and Vascular Surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jiang-Ming Sheng
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bai-Sui Feng
- Department of Gastroenterology, Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ke-Ping Peng
- Department of Otorhinolaryngology-Head and Neck surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Gui-Xiang Tian
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, China.,Research Center of Ultrasonography, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Cheng-Bai Liang
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming-Hui Liu
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hai-Qing Xie
- Department of Ultrasound, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qing Shu
- Department of Gastroenterology, First Affiliated Hospital, Shenzhen University, Shenzhen, China
| | - Yan Li
- Department of Gastroenterology, Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ping-Chang Yang
- Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
26
|
Sinha M, Zabini D, Guntur D, Nagaraj C, Enyedi P, Olschewski H, Kuebler WM, Olschewski A. Chloride channels in the lung: Challenges and perspectives for viral infections, pulmonary arterial hypertension, and cystic fibrosis. Pharmacol Ther 2022; 237:108249. [PMID: 35878810 DOI: 10.1016/j.pharmthera.2022.108249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
Fine control over chloride homeostasis in the lung is required to maintain membrane excitability, transepithelial transport as well as intra- and extracellular ion and water homeostasis. Over the last decades, a growing number of chloride channels and transporters have been identified in the cells of the pulmonary vasculature and the respiratory tract. The importance of these proteins is underpinned by the fact that impairment of their physiological function is associated with functional dysregulation, structural remodeling, or hereditary diseases of the lung. This paper reviews the field of chloride channels and transporters in the lung and discusses chloride channels in disease processes such as viral infections including SARS-CoV- 2, pulmonary arterial hypertension, cystic fibrosis and asthma. Although chloride channels have become a hot research topic in recent years, remarkably few of them have been targeted by pharmacological agents. As such, we complement the putative pathophysiological role of chloride channels here with a summary of their therapeutic potential.
Collapse
Affiliation(s)
- Madhushri Sinha
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Diana Zabini
- Department of Physiology, Neue Stiftingtalstrasse 6/V, 8010 Graz, Austria.
| | - Divya Guntur
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| | - Peter Enyedi
- Department of Physiology, Semmelweis University, Tűzoltó utca 37-47, 1094 Budapest, Hungary.
| | - Horst Olschewski
- Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Andrea Olschewski
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| |
Collapse
|
27
|
Zhang C, Wang X, Wang C, He C, Ma Q, Li J, Wang W, Xu YT, Wang T. Qingwenzhike Prescription Alleviates Acute Lung Injury Induced by LPS via Inhibiting TLR4/NF-kB Pathway and NLRP3 Inflammasome Activation. Front Pharmacol 2022; 12:790072. [PMID: 35002723 PMCID: PMC8733650 DOI: 10.3389/fphar.2021.790072] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Acute lung injury (ALI) is characterized by dysfunction of the alveolar epithelial membrane caused by acute inflammation and tissue injury. Qingwenzhike (QWZK) prescription has been demonstrated to be effective against respiratory viral infections in clinical practices, including coronavirus disease 2019 (COVID-19) infection. So far, the chemical compositions, protective effects on ALI, and possible anti-inflammatory mechanisms remain unknown. Methods: In this study, the compositions of QWZK were determined via the linear ion trap/electrostatic field orbital trap tandem high-resolution mass spectrometry (UHPLC-LTQ-Orbitrap MS). To test the protective effects of QWZK on ALI, an ALI model induced by lipopolysaccharide (LPS) in rats was used. The effects of QWZK on the LPS-induced ALI were evaluated by pathological changes and the number and classification of white blood cell (WBC) in bronchoalveolar lavage fluid (BALF). To investigate the possible underlying mechanisms, the contents of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein (MCP-1), interleukin-1β (IL-1β), interleukin-18 (IL-18), and immunoregulatory-related factors interferon-γ (IFN-γ) were detected by ELISA. Furthermore, the expression of Toll-like receptor 4 (TLR4), p-IKKα/β, IKKα, IKKβ, p-IκBα, IκBα, p-NF-κB, nuclear factor-κB (NF-κB), NOD-like receptor family pyrin domain containing 3 (NLRP3), cleaved caspase-1, pro-caspase-1, apoptosis-associated speck-like protein containing CARD (ASC), and β-actin were tested by Western blot. Results: A total of 99 compounds were identified in QWZK, including 33 flavonoids, 23 phenolic acids, 3 alkaloids, 3 coumarins, 20 triterpenoids, 5 anthraquinones, and 12 others. ALI rats induced by LPS exhibited significant increase in neutrophile, significant decrease in lymphocyte, and evidently thicker alveolar wall than control animals. QWZK reversed the changes in WBC count and alveolar wall to normal level on the model of ALI induced by LPS. ELISA results revealed that QWZK significantly reduced the overexpression of proinflammatory factors IL-6, TNF-α, MCP-1, IL-1β, IL-18, and IFN-γ induced by LPS. Western blot results demonstrated that QWZK significantly downregulated the overexpression of TLR4, p-IKKα/β, p-IκBα, p-NF-κB, NLRP3, cleaved caspase-1, and ASC induced by LPS, which suggested that QWZK inhibited TLR4/NF-κB signaling pathway and NLRP3 inflammasomes. Conclusions: The chemical compositions of QWZK were first identified. It was demonstrated that QWZK showed protective effects on ALI induced by LPS. The possible underlying mechanisms of QWZK on ALI induced by LPS was via inhibiting TLR4/NF-kB signaling pathway and NLRP3 inflammasome activation. This work suggested that QWZK is a potential therapeutic candidate for the treatments of ALI and pulmonary inflammation.
Collapse
Affiliation(s)
- Cai Zhang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinran Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng He
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jialin Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weiling Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan-Tong Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Abstract
Chloride transport across cell membranes is broadly involved in epithelial fluid transport, cell volume and pH regulation, muscle contraction, membrane excitability, and organellar acidification. The human genome encodes at least 53 chloride-transporting proteins with expression in cell plasma or intracellular membranes, which include chloride channels, exchangers, and cotransporters, some having broad anion specificity. Loss-of-function mutations in chloride transporters cause a wide variety of human diseases, including cystic fibrosis, secretory diarrhea, kidney stones, salt-wasting nephropathy, myotonia, osteopetrosis, hearing loss, and goiter. Although impactful advances have been made in the past decade in drug treatment of cystic fibrosis using small molecule modulators of the defective cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel, other chloride channels and solute carrier proteins (SLCs) represent relatively underexplored target classes for drug discovery. New opportunities have emerged for the development of chloride transport modulators as potential therapeutics for secretory diarrheas, constipation, dry eye disorders, kidney stones, polycystic kidney disease, hypertension, and osteoporosis. Approaches to chloride transport-targeted drug discovery are reviewed herein, with focus on chloride channel and exchanger classes in which recent preclinical advances have been made in the identification of small molecule modulators and in proof of concept testing in experimental animal models.
Collapse
Affiliation(s)
- Alan S Verkman
- Department of Medicine, University of California, San Francisco, California.,Department of Physiology, University of California, San Francisco, California
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
29
|
Yang X, An X, Wang C, Gao F, Lin Y, Chen W, Deng Q, Xu D, Li S, Zhang P, Sun B, Hou Y, Wu J. Protective Effect of Oxytocin on Ventilator-Induced Lung Injury Through NLRP3-Mediated Pathways. Front Pharmacol 2021; 12:722907. [PMID: 34733156 PMCID: PMC8558354 DOI: 10.3389/fphar.2021.722907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanical ventilation is an indispensable life-support treatment for acute respiratory failure in critically ill patients, which is generally believed to involve uncontrolled inflammatory responses. Oxytocin (OT) has been reported to be effective in animal models of acute lung injury. However, it is not clear whether Oxytocin has a protective effect on ventilator-induced lung injury (VILI). Therefore, in this study, we aimed to determine whether OT can attenuate VILI and explore the possible mechanism of this protection. To this end, a mouse VILI model was employed. Mice were pretreated with OT 30 min before the intraperitoneal injection of saline or nigericin and ventilation for 4 h, after which they were euthanized. Pathological changes, lung wet/dry (W/D) weight ratio, myeloperoxidase (MPO) activity, the levels of inflammatory cytokines [i.e., interleukin (IL)-1β, IL-6, and IL-18] in lung tissues and bronchoalveolar lavage fluid (BALF), and expression of NLRP3, Toll-like receptor 4 (TLR4), caspase-1, nuclear factor (NF)-κB, and GSDMD in lung tissues were measured. OT treatment could reduce pathological injury, the W/D ratio, and MPO activity in VILI mice. Our data also indicated that OT administration alleviated the expression of TLR4/My-D88 and the activation of NF-κB, NLRP3, and caspase-1 in lung tissues from the VILI mice model. Furthermore, OT also decreased the levels of IL-1β, IL-6, and IL-18 in the bronchoalveolar lavage fluid. Moreover, the OT administration may alleviate the activation of GSDMD partially through its effects on the NLRP3-mediated pathway. Collectively, OT exerted a beneficial effect on VILI by downregulating TLR4-and NLRP3-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Xiaomei Yang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Medicine, Cheeloo College of medicine, Shandong University, Jinan, China.,Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaona An
- Department of Anesthesiology, Zibo Central Hospital, Shandong University, Zibo, China
| | - Cheng Wang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Medicine, Cheeloo College of medicine, Shandong University, Jinan, China
| | - Feng Gao
- School of Medicine, Cheeloo College of medicine, Shandong University, Jinan, China
| | - Yicheng Lin
- Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjing Chen
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiming Deng
- Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongsheng Xu
- Department of Kidney Transplantation, The Second Hospital, Shandong University, Jinan, China
| | - Shengqiang Li
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Zhang
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baozhu Sun
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuedong Hou
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
30
|
Sirt3 Maintains Microvascular Endothelial Adherens Junction Integrity to Alleviate Sepsis-Induced Lung Inflammation by Modulating the Interaction of VE-Cadherin and β-Catenin. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8978795. [PMID: 34630854 PMCID: PMC8500765 DOI: 10.1155/2021/8978795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022]
Abstract
Inflammatory injury is a hallmark of sepsis-induced acute respiratory distress syndrome (ARDS)/acute lung injury (ALI). However, the mechanisms underlying inflammatory injury remain obscure. Here, we developed the novel strategy to suppress lung inflammation through maintaining microvascular endothelial barrier integrity. VE-cadherin is the main adherens junction protein that interacts with β-catenin and forms a complex. We found that lung inflammation was accompanied by decreased VE-cadherin expression and increased β-catenin activity in animal models and human pulmonary microvascular endothelial cells (HPMECs), illuminating the relationship among VE-cadherin/β-catenin complex, microvascular endothelial barrier integrity, and inflammation. Furthermore, we showed that the VE-cadherin/β-catenin complex dissociated upon lung inflammation, while Sirt3 promoted the stability of such a complex. Sirt3 was decreased during lung inflammation in vivo and in vitro. Sirt3 deficiency not only led to the downregulation of VE-cadherin but also enhanced the transcriptional activity of β-catenin that further increased β-catenin target gene MMP-7 expression, thereby promoting inflammatory factor COX-2 expression. Sirt3 overexpression promoted VE-cadherin expression, inhibited β-catenin transcriptional activity, strengthened the stability of the VE-cadherin/β-catenin complex, and suppressed inflammation in HPMECs. Notably, Sirt3 deficiency significantly damaged microvascular endothelial barrier integrity and intensified lung inflammation in animal model. These results demonstrated the role of Sirt3 in modulating microvascular endothelial barrier integrity to inhibit inflammation. Therefore, strategies that aim at enhancing the stability of endothelial VE-cadherin/β-catenin complex are potentially beneficial for preventing sepsis-induced lung inflammation.
Collapse
|
31
|
Do DC, Zhang Y, Tu W, Hu X, Xiao X, Chen J, Hao H, Liu Z, Li J, Huang SK, Wan M, Gao P. Type II alveolar epithelial cell-specific loss of RhoA exacerbates allergic airway inflammation through SLC26A4. JCI Insight 2021; 6:e148147. [PMID: 34101619 PMCID: PMC8410088 DOI: 10.1172/jci.insight.148147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/03/2021] [Indexed: 12/25/2022] Open
Abstract
The small GTPase RhoA and its downstream effectors are critical regulators in the pathophysiological processes of asthma. The underlying mechanism, however, remains undetermined. Here, we generated an asthma mouse model with RhoA–conditional KO mice (Sftpc-cre;RhoAfl/fl) in type II alveolar epithelial cells (AT2) and demonstrated that AT2 cell–specific deletion of RhoA leads to exacerbation of allergen-induced airway hyperresponsiveness and airway inflammation with elevated Th2 cytokines in bronchoalveolar lavage fluid (BALF). Notably, Sftpc-cre;RhoAfl/fl mice showed a significant reduction in Tgf-β1 levels in BALF and lung tissues, and administration of recombinant Tgf-β1 to the mice rescued Tgf-β1 and alleviated the increased allergic airway inflammation observed in Sftpc-cre;RhoAfl/fl mice. Using RNA sequencing technology, we identified Slc26a4 (pendrin), a transmembrane anion exchange, as the most upregulated gene in RhoA-deficient AT2 cells. The upregulation of SLC26A4 was further confirmed in AT2 cells of asthmatic patients and mouse models and in human airway epithelial cells expressing dominant-negative RHOA (RHOA-N19). SLA26A4 was also elevated in serum from asthmatic patients and negatively associated with the percentage of forced expiratory volume in 1 second (FEV1%). Furthermore, SLC26A4 inhibition promoted epithelial TGF-β1 release and attenuated allergic airway inflammation. Our study reveals a RhoA/SLC26A4 axis in AT2 cells that functions as a protective mechanism against allergic airway inflammation.
Collapse
Affiliation(s)
- Danh C Do
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yan Zhang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaojun Xiao
- Institute of Allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Jingsi Chen
- Children's Hospital, Chongqing Medical University, Chongqing, China
| | - Haiping Hao
- JHMI Deep Sequencing and Microarray Core Facility, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhigang Liu
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China.,Institute of Allergy and Immunology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Jing Li
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shau-Ku Huang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Wang Y, Liu YJ, Xu DF, Zhang H, Xu CF, Mao YF, Lv Z, Zhu XY, Jiang L. DRD1 downregulation contributes to mechanical stretch-induced lung endothelial barrier dysfunction. Am J Cancer Res 2021; 11:2505-2521. [PMID: 33456556 PMCID: PMC7806475 DOI: 10.7150/thno.46192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 12/05/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: The lung-protective effects of dopamine and its role in the pathology of ventilator-induced lung injury (VILI) are emerging. However, the underlying mechanisms are still largely unknown. Objective: To investigate the contribution of dopamine receptor dysregulation in the pathogenesis of VILI and therapeutic potential of dopamine D1 receptor (DRD1) agonist in VILI. Methods: The role of dopamine receptors in mechanical stretch-induced endothelial barrier dysfunction and lung injury was studied in DRD1 knockout mice, in isolated mouse lung vascular endothelial cells (MLVECs), and in lung samples from patients who underwent pulmonary lobectomy with mechanical ventilation for different time periods. Measurements and Main Results: DRD1 was downregulated in both surgical patients and mice exposed to mechanical ventilation. Prophylactic administration of dopamine or DRD1 agonist attenuated mechanical stretch-induced lung endothelial barrier dysfunction and lung injury. By contrast, pulmonary knockdown or global knockout of DRD1 exacerbated these effects. Prophylactic administration of dopamine attenuated mechanical stretch-induced α-tubulin deacetylation and subsequent endothelial hyperpermeability through DRD1 signaling. We identified that cyclic stretch-induced glycogen-synthase-kinase-3β activation led to phosphorylation and activation of histone deacetylase 6 (HDAC6), which resulted in deacetylation of α-tubulin. Upon activation, DRD1 signaling attenuated mechanical stretch-induced α-tubulin deacetylation and subsequent lung endothelial barrier dysfunction through cAMP/exchange protein activated by cAMP (EPAC)-mediated inactivation of HDAC6. Conclusions: This work identifies a novel protective role for DRD1 against mechanical stretch-induced lung endothelial barrier dysfunction and lung injury. Further study of the mechanisms involving DRD1 in the regulation of microtubule stability and interference with DRD1/cAMP/EPAC/HDAC6 signaling may provide insight into therapeutic approaches for VILI.
Collapse
|